Archives

Graves’ Disease and the Manifestations of Thyrotoxicosis

ABSTRACT

Graves' disease includes thyrotoxicosis, goiter, exophthalmos, and pretibial myxedema when fully expressed, but can occur with one or more of these features.

Graves' disease is a disease of "autoimmunity", but the final cause of autoimmunity remains unclear. A strong hereditary tendency is present. Inheritance of HLA antigens DR3, DQ 2, and DQA1*0501 predispose to Graves' disease. The abnormal immune response is characterized by the presence of antibodies directed against thyroid tissue antigens, including antibodies that react with the thyrotrophin receptor by binding to the receptor. Some of these antibodies act as agonists and stimulate the thyroid. The best-known of the antibodies is the serum factor first reported as  "LATS", now known as "TSAb".It has been reported in active Graves' disease that T- lymphocyte suppressor cell function is diminished and suppressor cell number is reduced. It is hypothesized that an abnormality in the control of autoimmune responses is present in this disease and leads to production of high levels of autoantibodies that may stimulate the thyroid or eventually cause thyroid damage and cell death.
The thyroid gland is hyperfunctioning in Graves' disease. The pituitary response to TRH is also suppressed. The gland is unusually responsive to small doses of iodide, which both block further hormone synthesis and inhibit release of hormone from the gland.
The incidence of Graves' disease is reported in recent studies to be 1 to 2 cases per 1,000 population per year in England. This rate is considerably higher than the rate of about 0.3 cases per 1,000 previously reported from this country. The frequency in women is much greater than it is in men.
The classic features of thyrotoxicosis are nervousness, diminished sleep, tremulousness, tachycardia, increased appetite, weight loss, and increased perspiration and signs are goiter, occasionally with exophthalmos, and rarely with pretibial myxedema. Physical findings include fine skin and hair, tremulousness, a hyperactive heart, Plummer's nails, muscle weakness, accelerated reflex relaxation, occasional splenomegaly, and often peripheral edema. Autoimmune vitiligo or hives may coexist in patients with Graves' disease.
The disease typically begins gradually in adult women and is progressive unless treated. Thyrotoxicosis can cause congestive heart failure. Mitral valve prolapse, atrial tachycardia and fibrillation are commonly caused by thyrotoxicosis.. Amenorrhea or anovulatory cycling is common in women, and fertility is reduced.Thyrotoxicosis in untreated cases leads to cardiovascular damage, bone loss and fractures, or inanition, and can be fatal. The long-term history also includes spontaneous remission in some cases and eventual spontaneous development of hypothyroidism, since autoimmune thyroiditis coexists and destroys the thyroid gland.

DEFINITIONS

Graves' disease is a syndrome characterized by hyperthyroidism, a particular ophthalmopathy, and pretibial myxedema. Rarely thyroid acropachy is associated. Usually thyroid enlargement, goiter, and excessive thyroid hormone action are the features of the illness, but the presence of all or any individual component fits a patient within the syndrome, and patients need not be hyperthyroid to have Graves' disease. The syndrome typically includes two major categories of phenomena. Those specific to Graves' disease, and caused by the autoimmunity per se, include the exophthalmos, thyroid enlargement and thyroid stimulation, and the dermal changes. The second set of problems is caused by the excess thyroid hormone. This thyrotoxicosis, or hyperthyroidism, does not differ from that induced by any other cause of excess thyroid hormone. The other causes of thyroid hormone excess are described in THYROIDMANAGER. Excess thyroid hormone causes a widespread disturbance in metabolism, since thyroid hormone effectively regulates the metabolic level in the body. For practical purposes, the two names, thyrotoxicosis, or hyperthyroidism, are used interchangeably.

Hyperthyroidism was first described in the English language by Caleb Perry (1755-1822), but it is the description by the Irish physician Robert Graves [1], to whom credit is usually attributed. The eponym Basedow's disease is often used on the European continent to recognize the description by Karl A. von Basedow (1799 - 1854).

GRAVES' DISEASE AS A DISEASE OF THYROID AUTOIMMUNITY

Graves' disease, Hashimoto's thyroiditis, and idiopathic thyroid failure are closely associated and in fact overlapping syndromes. Hashimoto's thyroiditis is typically characterized by thyroid enlargement and often underactivity. Idiopathic hypothyroidism is usually the result of Hashimoto's thyroiditis, and myxedema is the most advanced form of this illness. Of course hypothyroidism and myxedema can also be induced by other causes of thyroid hormone deficiency. These three syndromes of autoimmune thyroid disease (AITD) share immunological abnormalities, histological changes in the thyroid, and genetic predisposition. Patients can move from one or the other category, depending upon the stage of their illness. For example, an individual might first be observed with thyroid enlargement and positive antibody tests for anti-thyroglobulin or anti-TPO antibodies, and thus qualify as having Hashimoto's thyroiditis. At a later stage, this individual might become hyperthyroid and fit in the category of Graves' disease. Or, the patient with hyperthyroidism might have progressive destruction of the thyroid, or develope blocking antibodies, and become hypothyroid or ultimately develop myxedema.

The common features of the autoimmune thyroid diseases include the immune reactivity to specific thyroid antigens. We now know that patients with AITD have immune reactivity, both antibodies and cell-mediated immunity, directed to the TSH receptor, thyroid peroxidase (TPO), and thyroglobulin (TG) [2]. Antibodies also exist to megalin (the thyroid cell TG receptor)(3), to the thyroidal iodide symporter [4], and antibodies reacting to components of eye muscle and fibroblasts are present in sera of patients with Graves’ ophthalmopathy [5]. ( Table 10-1) The immune reactivity includes development of antibodies to these antigens, cell-mediated immune responses due to lymphocyte reactivity, and development of circulating antigen/antibody complexes [6], at least for some of the antigens. Patients with AITD also often develop other "organ specific" antibodies, including antibodies directed to gastric parietal cells in 50% of patients with Hashimoto’s thyroiditis [7]. Jenkins and Weetman have recently reviewed the evidence indicating an association of AITD with ACTH deficiency, Addison's disease, chronic hepatitis, celiac disease, DM-1, multiple sclerosis, myasthenia, PA, premature ovarian failure, primary biliary cirrhosis, vitiligo, RA, SLE, systemic sclerosis, urticaria, and angioedema(8). Patients with AITD may have antibodies, less frequently, to adrenal steroidogenic enzymes, ovarian steroidogenic enzymes, and components of the pituitary gland, thus qualifying for the Multiple Endocrine Autoimmune Syndrome [9]. In addition, up to 25% of patients with active Graves' disease have low level titers of antibodies to DNA, and occasionally have antibodies to liver mitochondria [10, 11]. Further evidence of ongoing autoimmunity in Graves' disease is the elevation of ICAM-1, and IL-6 and IL-8 cytokines seen in hyperthyroid patients [12, 13]. Anti-cardiolipin antibodies are present in increased incidence in patients with autoimmune thyroid disease, including Graves’ disease. However, these are not necessarily pathogenic and may be nonspecific markers of immune disregulation [14].

Table 1. Antibodies in Graves' Disease

 Elevated levels of TSAb, TBII, and (rarely) TSBAb

•  Elevated levels of anti-TPOAb ( 80%)

•  Elevated levels of anti-TGAb ( 50%)

•  Antibodies reacting to the Iodide Symporter and Pendrin protein

•  Antibodies recognizing components of eye muscle and/or fibroblasts

•  Antibodies to DNA

•  Antibodies to Parietal Cells (infrequent)

•  Antibodies binding to platelets

Graves' disease is associated statistically with a group of autoimmune diseases including pernicious anemia, vitiligo [15], alopecia [16], angioedema [17], myasthenia gravis [18], and idiopathic thrombocytopenic purpura [13]. A weak association is probably present with rheumatoid arthritis and systemic lupus erythematosus [19]. Graves' disease is an example of an organ specific autoimmune disease, and appears not to be statistically more common among individuals who have, dermatomyositis, or scleroderma [20].

Up to 90% of patients with Graves' disease have antibodies directed to the "microsomal antigen" in the thyroid, known now to be thyroid peroxidase [21-25]. A lower proportion, approximately 50%, have antibodies directed against thyroglobulin [25]. Rarely patients have antibodies directed against T4 or T3 [23]. These antibodies are very similar to those present in Hashimoto's thyroiditis and idiopathic myxedema. Peripheral blood mononuclear cells [26], thyroid lymphocytes [27], and lymph node lymphocytes demonstrated cell-mediated immunity to TG, TPO, and TSH-R [28,29], and also to specific peptide epitopes of TSH-R,TPO and TG [30-35]. The functional consequence of having TG antibodies is uncertain, but they do not appear in general to cause thyroid cell destruction. TG/anti-TG immune complexes are rarely deposited in the kidney basement membrane of the glomeruli and can, in extremely rare circumstances, produce disease [36-38]. Anti-TPO antibodies are not known to play a role in Graves' disease, although they are thought possibly to be cytotoxic and function in the pathology of Hashimoto's thyroiditis [38]. Presumably TPO antibodies could similarly cause cytotoxicity in Graves' disease. About 1/3 of patients with autoimmune thyroid disease have ANA antibodies, and the ANA+ patients may also have antiRo, anti-La, anti-dsDNA , and anti-cardiolipin antibodies, and up to 10% have Sjogren's Syndrome.(27.1).

TSH-Receptor Antibodies

The antibodies of central importance in Graves' disease are those directed against the TSH receptor on the thyroid cell membrane. Protein factors in the circulation, thought to play a role in Graves' disease, have been described for more than five decades. Serum factors were described which produce exophthalmos in experimental models including fish and guinea pigs, and were given the eponym Exophthalmos Producing Substance [40,41]. For a time this material was thought to be a modified TSH molecule.

The first clear evidence of a circulating factor that could induce thyroid hyperactivity came from studies by Adams and Purves , who showed the presence of a factor in human serum that could stimulate the thyroid of guinea pigs [42]. Ultimately they found that infusion of this material into a human stimulated release of hormone from the thyroid [43](Figure 10-1). Because of the time course of its action being longer than TSH, this material was labelled Long Acting Thyroid Stimulator, or LATS. It was subsequently shown to be a gammaglobulin, and thus began the entirely new concept of an autoimmune disease due to stimulation of the thyroid by an antibody to an antigen in the thyroid, that mimicked the action of the natural stimulator, TSH. Nearly three decades later, the antigen to which this antibody was directed was identified as the thyroid cell surface protein receptor for TSH (TSH-R) [44,45].

Figure 1. Stimulation of thyroid secretion by LATS-P, a form of TSAb. The subject's thyroid iodine was labeled by administration of 131I, and serial observations were made on the appearance of 131I-labeled hormone in blood (ordinate) over one month (abscissa). An infusion of 280 ml control plasma had no effect, but 280 ml plasma from a patient with Graves' disease caused a marked stimulation of secretion of hormone from the thyroid. (D.D. Adams et al., J. Clin. Endocrinol. Metab., 39:826, 1974. Used with permission of the authors). Of interest, the subjects initials were D.A.

Over the past three decades, an enormous literature has grown around the identification, quantitation, and pathophysiologic importance of these anti-receptor antibodies [46-48]. The antibodies can be classified into at least three general types. Thyroid stimulating antibodies (TSAb, sometimes TSI) interact with the TSH receptor in a positive functional manner and initiate the adenyl cyclase function and the phospholipase A2 function of the receptor, causing all aspects of thyroid stimulation [49-51]. Functionally, this is probably identical to the effects induced by TSH itself. Other antibodies can interact with the receptor in a slightly different manner, presumably by binding to different epitopes on the receptor, and can block the binding of TSH to the receptor while not themselves stimulating function. These antibodies are known as thyroid stimulation blocking antibodies, or TBAb [52-55]. A third set of antibodies can bind to the receptor but neither stimulate nor inhibit its function. These are known as thyrotrophin binding immunoglobulins. They are commonly recognized by assays which detect their ability to interfere with the binding of TSH to the receptor, and are identified as TRAb, or TBII [53]. Probably all patients with Graves' disease have a mixture of all of these antibodies. If TSAb predominate, thyroid stimulation occurs and, if the activity is sufficient, the patient may become hyperthyroid and be characterized as having Graves' disease. If the antibodies block the action of TSH, they may induce hypothyroidism, in which case the patient might be characterized as having Hashimoto's thyroiditis or idiopathic myxedema 

TSAb, TBAb, and TRAb

TSAb are usually identified by an assay which quantitates the ability of the antibodies to stimulate the adenyl cyclase function of the TSHR membrane receptor. Either thyroid cells or thyroid cell membranes can be used, and the cyclic AMP produced by this stimulation is quantitated by a radioimmune assay [49]. A cyclic AMP responsive luciferase construct has been stably introduced into CHO cells, allowing a sensitive luminescent assay for thyroid stimulating antibodies with the capability of high throughput suitable for use in general laboratories [50].Assays which measure DNA synthesis of the thyroid cells, or some other aspect of cell growth such as incorporation of thymidine, may or may not measure the same type of antibodies. Antibodies having this action are called thyroid growth stimulating antibodies, to indicate a potential difference [52], and are reported to be present in sera of patients with multinodular goiter. TBAb are measured in the same kind of assay as are TSAb. However, in their assay, a basal level of stimulation is obtained by using bovine or human TSH, and then the ability of the TBAb antibody to interfere with this stimulation is quantitated as "blocking activity" [53]. TRAb are typically measured by their ability to interfere with the binding of radiolabelled TSH to thyroid cells or thyroid cell membranes. These IgGs can be of several subclasses, and have a restricted clonal but not monoclonal origin [56]. They can bind to certain animal TSH-R molecules and to TSH-R (apparently) present on fat cells [57]. Although some studies suggest limited clonality of the anti-receptor antibodies, several studies indicate that both types of light chains are present, and the antibodies may be of a mixture of IgG1, IgG2, IgG3 and IgG4. Antibodies with lower affinity can be found in normal patient sera.

TSAb mediate the thyroid hyperactivity and hypersecretion characteristic of Graves' disease. Presumably low levels of TSAb can stimulate the thyroid in a way that replaces TSH stimulation, and can make the thyroid non-suppressible by administered thyroid hormone, but not cause overproduction of hormone. However, when TSAb reach a certain level of function, they cause an increased secretion of thyroid hormone and produce hyperthyroidism.

The TSH receptor is formed as one polypeptide chain and inserted into the thyroid cell plasma membrane. It undergoes a processing that is reminiscent of that occurring with insulin. A segment of 30 or more amino acids is cut out of the receptor at approximately residue 320, forming a two peptide structure with the chains held together by disulfide bonds. It is thought that both the intact and the processed receptor are functional. The processing of the receptor is thought to involve a matrix metalloprotease-like enzyme cleaving the 120 kDa precursor to form the heterodimeric receptor. Subsequently, reduction of the disulfide bonds by a protein disulfide isomerase may separate the two molecules and lead to shedding of the “alpha” subunit. It is an interesting concept that shedding of the alpha subunit might be intimately related to onset of autoimmunity against the TSH receptor. Shedding of the receptor is augmented by TSH stimulation of thyroid cells (58). The amino-terminal ectodomain of the human TSH receptor has been expressed on the surface of CHO cells as a glycosylphosphatidylinositol-anchored molecule. This material can be released from the cells and is biologically active in that it binds immunoglobulins from serum of patients with Graves’ disease, and displays saturable binding of TSH (46), indicating that all of the "immunologic information" related to production of antibodies resides in the extracellular portion of TSH-R.

The initial bioassay for TSH developed by Adams [42], and then by MacKenzie [59], could quantitate TSAb (or LATS as it was then known) in up to 60% of patients with active Graves' disease. Recent assays measuring thyroid stimulation by cyclic-AMP formation can detect TSAb in over 90% of patients with active thyrotoxicosis [60]. Newer assays being described may increase this to near 100%. The presence of TSAb is thus presumed to be characteristic of active Graves' disease, and if the thyroid can respond, induces hyperthyroidism. The natural course of such antibody action on the thyroid in the untreated state is not usually observed at present. However, the stimulating antibodies are typically associated with other antibodies, and cell-mediated immunity, which damage the thyroid. In time, if the patient survives, the thyroid may be destroyed, or develop blocking antibodies, and the patient may become hypothyroid. TSH-R antibodies are found in patients with Hashimoto's thyroiditis, infrequently in patients with toxic multinodular goiter, and rarely in "normal" subjects [61].

During antithyroid therapy TSABs tend to decline, and if present in significant concentration at the end of a period of therapy, the patient rarely enters remission [62]. Similarly, after surgery, TSAb tend to decline if the patient enters a euthyroid state [63]. After radioactive iodide therapy, TSAb are stimulated to increased levels during several months or a year, probably because of release of antigens [64,65]. TSAbs gradually return to lower levels during the subsequent years. It is also possible that, during antithyroid drug therapy, some sort of immune modulation occurs and the predominant stimulating antibodies are replaced by antibodies which have binding or blocking activity. The specific epitopes to which the thyroid stimulating antibodies bind on the TSH receptor have not been identified. There is evidence that TSAb bind to sequences in the amino terminal portion of the extracellular domain, while those with blocking activity tend to bind to sequences at the carboxy terminal portion nearer the plasma membrane [66].

Lymphocyte reactivity to TSH-R

Lymphocytes of patients with Graves' disease are reactive to TSH-R , as shown by their proliferation during in vitro incubation with TSH-R in vitro [67], and to peptides derived from the TSH receptor. We have shown that patients' lymphocytes react to peptide sequences in TPO (AA110-129, 211-223, 842-861, and 882-901), as well as from TSH-R (AA44-62, 158-176, 237-252, and 248-263)[ 32,68-70]. Thyroid stimulating hormone receptor specific T cells have also been developed from thyroid tissue of patients with Graves’ disease [71]. TSHR peptides  (GLKMFPDLTKVYST) and  (ISRIYVSIDVTLQQLES) that have aspartic or glutamic acid in in position four of their binding motif are partiularly active inducing Tcell responses in Graves’ disease and immunized animal T cells, and are probably important in developing the immune response to TSHR (72).

It is probable that immunity to TSH receptor plays a direct role in the development of Graves’ ophthalmopathy, probably mediated by immune cell secretion of imflammatory cytokines.. Crisp et al observed immunoreactive TSH-R in samples of normal and orbital fat. Up to 5% of orbital preadipocytes displayed TSH-R reactivity. Differentiation of preadipocytes into adipocytes was induced by TSH stimulation, and on differentiation, more of the adipocytes displayed TSH-R reactivity, and also cyclic-AMP production after TSH stimulation [73-74].Bell et al also found TSH-R mRNA in both orbital and abdominal adipose tissue samples, and TSHR protein in these tissues. TSH activated preadipocytes. In addition to the relation to Graves’ ophthalmopathy, these authors suggest that TSHR signalling may be important in adipose tissue development [75]. Haraguchi et al report that TSH causes proliferation and inhibits differentiation of rat preadipocytes, again supporting the idea that TSH-R may be an important regulator of this process in animals and possibly in man , at least in adipocytes in the orbit[75]. Immunity to TSH-R is believed to lead to production of cytokines in the orbital tissue, which actually mediate the inflammatory process. Hiromatsu et al investigated cytokine profiles in eye tissue and found that Th1 cytokines such as IFNγ, TNFα, IL-1β, and IL-6 were primarily present in eye muscle whereas IL-4 and IL-10 were detected in many samples of orbital fat. Thus both Th1-like and Th2-like immune reactivity may play a role, although it may differ in the involved tissues [76]. Wakelkamp et al found that serum concentrations of sIL-2R, IL-6, sIL-6R, TNFα, and sCD30 were elevated in Graves’ ophthalmopathy patients compared to controls. They also conclude that both Th1 and Th2 type cytokines are elevated in these patients [77].

The role of TSH-R antigenic stimulation is also suggested by triggering of Graves’ disease by treatment of non-toxic goiter with large doses of 131I, which releases antigens from the thyroid, with some patients developing typical ophthalmopathy [78].

Figure 1a-Schematic representation of TSH-R endocytosis by antigen presenting cells, subsequent proteolysis of the receptor into 9-20 amino acid fragments, their “presentation” in DR molecules to T cells, which help B cells produce antibodies, leading to production of TSAb. (Figure courtesy of Hidefumi Inaba, MD)

The importance of immunity to the TSH-R as the basic effector in Graves' Disease is strongly supported by the development of animal models. Ludgate and coworkers immunized outbred mice by injection of a plasmid expressing TSH-R, and the mice developed some features of Graves’ disease [79]. Nagayama and associates and others have developed this model immunizing mice with an adnovirus or plasmid expressing the A-subunit of TSHR, developed TSAb and elevated T4 levels, and in some studies  evidence of ocular pathology(81,82). In line with the role of HLA DRB1*0301 fostering Graves in humans(see above), mice made transgenic for expression of this human HLA protein are more susceptible to this induction of "Graves' disease" than are mice transgenic for other HLA proteins(72,80).

Auto-antibodies to IGF-1 Receeptor-??

A role for antibodies binding to and stimulating the IGF-1 receptor has been proposed. IgG isolated from the sera of patients with Graves' disease (GD-IgG) provoked the synthesis of hyaluronan in orbital fibroblasts. The effect of GD-IgG was reproduced by IGF-I, and appeared to be mediated through the IGF-I receptor. In contrast to the effects in GD fibroblasts, cultures derived from donors without known thyroid disease fail to respond to GD-IgG or IGF-I. The observation that hyaluronan production is induced by GD-IgG in fibroblasts suggests that the IGF-I receptor and its activating antibodies could represent a pathway through which important pathogenic events in thyroid-associated ophthalmopathy are mediated (83). B-cells from patients with Graves” are reported to aberrantly express the IGF-1-R, suggesting that these antibodies could stimulate their own production by B cells (84 ). IGF1R antibodies exist in sera from about 10%  of “normal” subjects, and  a similar  percent of GD patients, and are stable over time, indicating they have no unique role in GD(85  ). Gershengorn et al demonstrated that the monoclonal hTSHR Ab, M22, stimulated HA secretion by GO fibroblasts/preadipocytes and this involved cross talk between TSHR and IGF-1R(86). This cross talk releys on TSHR activation rather than direct activation of IGF-1R and leads to synergistic stimulation of HA secretion. It currently seems unlikely that specific anti-IGFR antibodies play an important role  in GO. However IGF1,PDGF-AB and BB augment the action of TSH and TSHR antibodies to produce hyaluronan by orbital fibroblasts (87), supporting the idea  that coincident stimulation of TSHR and IGFR on fibroblasts of adipocytes may be important.

CAUSES OF GRAVES' DISEASE

Since Graves' disease is accepted as a disease syndrome induced by autoimmunity to the thyroid, the question of cause resolves into why autoimmunity to the thyroid is present. It is not clear that any other "cause" of Graves' disease is present, other than disordered immunity. There is, for example, no evidence that the thyroid or its protein antigens are initially abnormal [88]. Contemporary understanding is that the process involves a variety of factors allowing self-reactivity to occur. ( Table 10-2) While our immune system is designed to prevent self-reactivity, to some extent, very low levels of self-reactity are normally present [2]. Presumably genetic and environmental factors interact to augment this immunity, from a low and physiologically unimportant level, to a degree that causes a disease state. Several such factors can be identified with some certainty, and others have been suggested and will be noted. It always remains possible, when a specific individual cause is not known, that ultimately one single cause of Graves' disease will be discovered. Current ideas suggest this is not the case.

Table 2. Possible Factors in  the Immunological Etiology of Graves' Disease

Persistence of some autoreactive T cells and B cells (failure of negative selection)

•  Inheritance of specific HLA, CTLA-4,  and many other immune-response related genes

•  Re-exposure of antigens by thyroid cell damage

•  Reduced or dysfunctional regulatory T cells

•  Cross-reacting epitopes on environmental and thyroid antigens

•  Inappropriate HLA-DR expression

•  Mutated T or B cell clones

•  Activation of T cells by polyclonal stimuli

•  Stimulation of the Thyroid by Cytokines

Environmental Factors- Recognised “Causes” of Graves’ disease

Considerable information has accumulated about factors in the environment that can  induce Graves' disease. Damage to the thyroid, by radiation or ethanol injection, with liberation of antigens, has been noted above. Induction of thyroid autoimmunity, including Graves disease, following therapeutic use of IL-1alpha, IL-2, IFN alpha and gamma, CAMPATH, and drugs used in HIV treatment, are noted below. Cigarette smoking increases risk for GD about 2 fold, while alcohol use and physical activity seem unrelated, and obesity decreases risk(89).

Thymic Selection of Lymphocytes

Lymphocytes develop from precursors present in the bone marrow and mature in the thymus, where they undergo progressive maturation and selection. Lymphocytes which fail to recognize endogenous HLA molecules undergo negative selection, as do those which strongly react with endogenous epitopes presented by HLA molecules [90,91]. In this process, 95 - 98% of all lymphocytes developing in the thymus undergo apoptosis. Spitzweg et al report that NIS, TSH-R, TPO, and Tg-RNAs are present and processed to immunoreactive peptides in the human thymus, and other groups have reported similar findings (92,93). These data suggest that pre-T lymphocytes may be educated in the thymus to recognize thyroid-related epitopes, and thus to generate self-tolerance against these thyroid-related antigens. Expression of these thyroid antigens in the fetal thymus is under control of the AIRE gene, and absence of this gene leads to a fatal poly-autoimmune disease. In Downs syndrome , despite having 3 copies of the AIRE gene, expression of thyroid antigens in the thymus is reduced, and this is thought to cause the elevated incidence of autoimmunity in Down’s (94 ).Clearly this process in imperfect, since cells reacting with these antigens are present in the peripheral blood of normal patients and those with autoimmune thyroid disease [92]. Presumably, in a developmental process designed to provide the maximum repertoire of lymphocytes, some lymphocytes which weakly recognize autologous antigens in the context of autologous HLA are allowed to persist in the circulation. Whether this varies from person to person and is involved in the selection for Graves' disease is unknown.

Molecular Mimicry

A persistent theory on the etiology of autoimmune diseases is that exposure to a particular peptide epitope in an environmental antigen might develop immune reactivity to an amino acid sequence identical to that present in an human endogenous antigen such as TSH receptor, TPO, or TG. Through this molecular mimicry, exposure to a virus or bacteria could produce heightened immune reactivity to a component of the body. This sequence is believed to play a role in rheumatic fever and glomerulonephritis. No certain examples of this sequence have been shown for thyroid disease. However, there is some evidence that proteins present in a common intestinal parasite, Yersinia Enterocolitica, may induce antibody reactivity to TSH receptor [95,96]. These proteins actually appear to be coded for by plasmids which live within the pathogenic bacteria [97], and a specific sequence in the plasmid (DALYGNVTS) is  similar to a sequence (198-DAFGGVYS-205) in TSHR. A higher proportion of patients with Graves' disease have been infected with this bacteria than people who do not have Graves' disease [95], exposure to the bacteria can induce TSH receptor antibodies [96], and TSH appears to bind to a molecule on the bacteria [98]. There is also evidence for infection of the thyroid by foamy viruses [99, (although this is disputed) , and there is clear evidence that autoimmunity to the thyroid is induced by infection with the HLTV1 virus, which causes lymphocytic leukemia [100]. Whether this is due to molecular mimicry of the virus, viral damage to the thyroid, or stimulation through another mechanism such as cytokine secretion, remains uncertain. While molecular mimicry remains tantalizing, the factual evidence for its role in the pathogenesis of Graves' disease is minimal.

Another environmental factor that may have an effect on the development of autoimmune thyroid disease is H. pylori infection of the gastric mucosa. One study reports that up to 85% of patients with autoimmune atrophic thyroiditis have H. pylori infection, and it has been suggested that H. pylori antigens may be involved in the development of autoimmune thyroid disease (101). Interestingly, rosacea is also associated with a high prevalence of H. pylori infection, and eradication of H. pylori leads to an improvement in symptoms of gastritis and of rosacea (102).

Thyroid Injury and Antigen Release

It is definite that certain types of injury to the thyroid are followed by the development of thyroid autoimmunity, including Graves' disease. In fact this is one of the few proven cause of Graves' disease. We recognized three decades ago that radiation to the thyroid in young people was followed by a higher incidence of positive thyroid antibody tests [103]. Hancock and associates have reported a significantly increased risk of Graves' disease, Hashimoto's thyroiditis, and Graves’ophthalmopathy associated with radiation to the neck for Hodgkins disease [104]. The incidence of thyroid autoimmunity is elevated among children and adolescents radiated by the Chernobyl explosion ( 105) Radioactive iodide treatment for toxic multinodular goiter and ethanol injection for cure of toxic thyroid nodules have both been followed by the development of autoimmune Graves' disease [106,107]. Over 1 % of patients treated with RAI for autonomous thyroid disease develop Graves hyperthyroidism after therapy, and the incidence is 10x greater if anti-TPO antibodies are present (108). Thus, in this marvelous but unintentional manner, it has been demonstrated that release of thyroid antigens may add a significant stimulation to a latent low level of thyroid autoimmunity, causing the development of clinically important Graves disease, including ophthalmopathy. Whether viral injury, as in the case of HLTV1, plays a similar role is uncertain, although it has been shown experimentally that Reo virus infection of a neonatal mouse can induce thyroiditis and thyroid autoimmunity [109].

Alterations in immune function and  cytokines

(See discussion of Tregs below) Administration of cytokines IFN-alpha, IL-2, and GM-CSF in chemotherapy can augment AITD, or in some cases appear to induce it de novo. These agents may act to magnify latent immunity, and a direct action on the thyroid cell may also be involved [110-112]. Depletion of circulating lymphocytes is used in therapy of Multiple Sclerosis. In a group of 34 patients so treated (using CAMPATH),a humanized monoclonal antibody), one-third developed Graves' Disease within 6 months during recovery from T cell depletion. Patient’s stopping immunosuppressive treatment of TIDM may have “re-bound” development of Graves’ disease. These treatments may deviate the immune system from a TH1 to a TH2 type of response [113]. Most likely these therapies reduce the number  of Tregs, or alter the balance between effector cells  (Th17, Th 23) and regulatory T cells(114).
Cells expressing the pro-inflammatory cytokines IL-17, IL-22, and IL-23R are increased in blood and thyroid of patients with AITD, including Graves” (114).

Onset or worsening of antithyroid autoimmunity and thyroid dysfunction have been reported to occur during treatment with Interferon-α for chronic hepatitis, or Interferon-β for multiple sclerosis. Interferon-α treatment of patients with chronic hepatitis due to hepatitis C virus is associated with the development of primary hypothyroidism, Graves’ hyperthyroidism, and destructive thyroiditis, and is especially prevalent in women (relative risk of 4.4), and in the presence of existing TPO antibodies (relative risk of 3.9)(116). However, one study of this problem in a large group of patients treated for multiple sclerosis found no evidence of a trend to development of thyroid dysfunction during Interferon-β treatment (117).

Genetic Factors

The increased incidence of Graves' disease in certain families [118,119]and in identical twins [120,121] has for decades indicated a powerful genetic influence on development of the disease. Studies of pairs of twins suggest that the genetic factors account for 79% of the liability to the development of Graves’ disease, whereas environmental factors account presumably for the remainder (121).A representative sample of healthy twin pairs was identified through the Danish Twin Registry; 1372 individuals, divided into 283 monozygotic (MZ), 285 dizygotic same sex (DZ), and 118 opposite sex twin pairs were investigated. Serum TPOab and serum Tgab were measured. Proband-wise concordance and intraclass correlations were calculated, and quantitative genetic modelling was performed. Genetic components (with 95% confidence intervals) accounted for 73% (46-89%) of the liability of being thyroid antibody positive. Adjusting for covariates (age, TSH and others), the estimate for genetic influence on serum TPOab concentrations was 61% (49-70%) in males and 72% (64-79%) in females. For serum TGAb concentrations, the estimates were 39% (24-51%) and 75% (66-81%) respectively (122). Early markers of thyroid autoimmunity appear to be under strong genetic influence.

The first genetic factor to be associated with Graves' disease was HLA-B8 [123], a Class I major histocompatibility component (MHC). Inheritance of this gene, expressed on the surface of antigen presenting cells, was found to confer increased risk of getting Graves' disease. Subsequently, this relation was found to be more specifically with an MHC Class II molecule, HLA-DR3 [124,125]. Inheritance of this gene increases the risk of Graves' disease up to 5.7-fold. Our laboratory demonstrated that the HLA molecule DQA1*0501 was also closely and independently associated with the risk of getting Graves' disease [126,127]. In contrast, inheritance of HLA DRbeta 1*07 appears to be protective [128].

The reason HLA genes are associated with Graves' disease seems now to be clear, although the exact pathway is less certain. These molecules exist as dimers on the surface of antigen presenting cells. In the initiation of an immune response, the antigen presenting cell displays a specific epitope complexed in a DR protein. Recognition of this bi-molecular complex by the T cell receptor leads to stimulation of the T cell. In contrast to the possible 10-7 or higher specificities present on individual T cell receptors, the spectrum of HLA molecules is much more restricted. There are between 50 and 100 different DR molecules, and a much smaller number of DQ and DP molecules, all coded on Chromosome 6, in the human genome [129]. Each human has genes coding for two DR, two DQ, and two DP molecules. Of these, the DR are most abundantly expressed and most important. The HLA molecules exist as dimers on the surface of antigen presenting cells, and their extracellular domains form a structure that can be compared to a hot dog bun, into which the peptide epitope is cradled much like a hot dog in a hot dog bun. (Figure 10-1.1, below) This combination, the DR molecule and its enclosed amino acid epitope of 9 - 20 amino acids, constitutes the structure which is seen and recognized, or not, by the receptor on a T cell receptor.

The amino acid sequences of the DR molecule determine the shape of the antigen presenting cleft, and peptides formed from protein antigens fit into the cleft with greater or lesser affinity, depending upon how well their three dimensional structure fits into the three dimensional structure of the antigen presenting cleft of the HLA molecule [130]. The net effect of this is that certain DR molecules are better able to present certain epitopes. DR molecules which best fit epitopes derived from the TSH receptor, for example, are most effective in presenting the epitope to the T cell to induce immunity. This same recognition sequence is involved in selection of T cells in the thymus and determines whether the T cells are destroyed or allowed to mature(90). Thus the matching of the DR molecule with the structure of the TSHR epitopes, or other thyroid epitopes, plays an important role in determining the development of autoimmunity. Sawai and DeGroot studied the binding of TSH receptor peptide epitopes to the DRB1*0301 molecule known to be associated with Graves’ disease. Epitopes which induce reactivity of T cells from patients with Graves’ disease bound with medium  affinity, whereas epitopes which did not stimulate T cells bound with very low affinity(131). De Groot and Inaba provided a specific molecular explanation for this role of DR3 is promoting Graves’ disease. They found that TSH-R-ECD epitopes with aspartic or glutamic acid (D/E) in positions 71 and 74 in the DR sequence  (important in defining the shape and charge of pocket 4 on the surface of the DR molecule)  bind more strongly to DRB1*0301 than epitopes that are D/E- and are more stimulatory to GD patients' peripheral blood mononuclear cells and to splenocytes from mice immunized to hTSH-R (72). This effect is due to the presence of arginine at position 74 in the β-chain of the HLA-DR molecule, giving a strong positive charge to pocket 4. Thus epitopes with D or E at specific positions in their motif are important in immunogenicity to TSH-R due to their favored binding to HLA-DR3, thus increasing presentation to T cells.

These data support the concept that the ability of specific peptides to bind in the antigen binding cleft of the HLA molecule is the reason for the association between the HLA Class II DR3 or DR5 molecules and the development of Graves’ disease [2]. Individuals who have DR3 antigen, or HLA-DQA*0501, tend to develop Graves' disease [124-126]. Those who have DR5 tend to develop Hashimoto's thyroiditis [132]. This correlation does not necessarily hold perfectly true from one human race to another, but there is a general similarity that suggests great importance for this relationship. However, the relationship between DR gene inheritance and Graves' disease is such that it can account for about a 2 - 5-fold increment in risk, which is certainly not enough to explain the marked increase in risk for Graves' disease seen in many families.

The importance of the HLA-DRβ1*0301 gene in autoimmune thyroid disease has been demonstrated by using transgenic mice. Transgenic mice expressing DR3 were susceptible to induction of thyroiditis following immunization with thyroglobulin, whereas animals transgenic with DRβ1*1502 (DR2) were resistant(133). Pichurin et al found that transgenic mice that express HLA-DR3 without their own murine MHC are prone to develop TSH receptor antibodies after vaccination with TSHR-DNA in a plasmid, whereas control mice that were HLA DQ6b transgenic did not develop the antibodies. Some of the sera recognized a linear peptide sequence present in the amino terminus of the TSHR (134).

Figure 1.1. In this remarkable x-ray crystallographic study, an HLA Class I molecule is seen from above. The two interlocking subunits form an antigen binding cleft into which the peptide epitope must fit and remain if it is to be recognized by the T cell receptor(a). In (b), fortuitously, a peptide epitope is found occupying the cleft, fitting like the hot-dog in a bun.

 

Figure 1.2- Schematic diagram of how a peptide, such as one derived from TSH-R, can fit into the 9 “pockets” formed on the surface of a DR molecule by the peptides in the alpha and beta chains. The amino acids fit into a groove with one aspect of their structure facing the DR molecule, and the other facing outward and becoming, with the surface of DR, the structure that is recognized by specific T cells.

CTLA4:

A second gene was found to be related to the propensity to develop Graves' disease, and this gene also is involved in immune responses [135,136]. When an immune reaction begins, the "first signal" is through the recognition by the T cell receptor of its cognate epitope presented in an HLA molecule. However, if only the first signal is received by a T cell, the T cell tends to be turned off or “anergized”. In order for a progressive immune response to recur, there must be a "second signal" provided by one of several adhesion molecules which exist on the APC and T cell, and which tend to augment the affinity of the interaction [137]. Of these, one of the most important is "B7", which exists in two forms, B7.1 and B7.2, present on the surface of APCs. These molecules interact with their cognate receptors on the T cell, CD28 for B7.1 and CTLA4 for B7.2. In many situations interaction between B7.1 and CD28 give a positive stimulus to growth of the T cell, whereas interaction with CTLA4 provides a negative signal [138]. CTLA4 exists as a gene with several isoforms. These are due to a polymorphism in the leader sequence, and an AT dimer polymorphisms in the 3’-untranslated region of exon 3, which are also closely linked with the polymorphism in exon 1 [139]. It has been found that inheritance of the 106 base pair AT polymorphism is associated with a greater incidence of Graves' disease, especially in males [135,136]. The G (alanine ) position 49 allele was found to be linked to Graves' Disease by Heward et al [140] and Vaidya et al found in a linkage study that inheritance of a specific CTLA-4 allele along with MHC allele was responsible for 50% of the genetic influence in Graves' disease [141] CTLA gene polymorphism studies indicate that the G allele, associated with the development of Graves’ disease, also influences higher production of TPO and Tg antibodies [142].

Kouki and De Groot investigated the relation of the CTLA-4 alleles to proliferation of T cells in patients with Graves’ disease and Hashimoto’s thyroiditis. They found that T cells from all subjects that have the G polymorphism, including normal controls, proliferate to a greater degree than do lymphocytes bearing the CTLA-4 A polymorphism. This is presumed to give individuals carrying the G polymorphism a mild but important greater propensity for development of a functional auto-reactive lymphocyte clone [143].

Interestingly, the HLA association suggests a relationship to disease specificity, since it has to do with the presentation of specific antigen epitopes, whereas the CTLA-4 polymorphism appears to be a general phenomenon, allowing one population group to have augmented lymphocyte proliferation, but is not specifically related to the disease. These observations also fit with the concept that development of Graves’ disease is mediated by a set of genes rather than one specific gene. It is reasonable that a variation in the function of the CTLA-4 gene makes it less effective, as a suppressive signal controlling autoimmunity. The genetic effects of DR genes and CTLA-4 interact. Specifically, it has been found that the positive effect of CTLA-4 predisposition mitigates in part the negative effect of DRB1*0701, but does not interact with the positive influence of DRB1*0301(33)

Numerous other gene polymorphisms  have been reported to be associated with GD. Very likely these genes all provide a real but small increment in the chance of developing GD. It has been indicated [144], and denied [145], that an association exists between a specific polymorphism of the TSH receptor (PRO52THR), and susceptibility to Graves' disease. Alleles of intron 7 of the TSH-R gene were found associated with GD in Japanese patients(146). Linkage to the TSH-R gene has recently been confirmed by Dechairo et al [147]. The TG gene has been linked to Graves and other AITD, but to date evidence for this relation is uncertain.  A Vitamin D receptor exon 2 initiation codon polymorphism has been associated with Graves’ disease in a Japanese population. A similar association has been reported with IDDM and multiple sclerosis [149]. Vitamin D and its receptor are involved in control of autoimmunity, so an association is not surprising, but the mechanism remains unknown. Inheritance of specific V genes coding for immunoglobulins may carry the same kind of risk. Several possible genes linked to Graves’ disease or autoimmune thyroid disease have been found by linkage studies, including one recently described at a locus on chromosome 18q21 that is also associated with IDDM. (148).
Linkage studies of Graves’ family members have suggested the susceptibility locus on chromosome 20q11, which has been named GD-2 by Davies et al, is related to the gene CD-40, expressed on B cells and other immune cells. Recently this group identified a polymorphism in the Kozak sequence of the CD40 gene at position –1 from the translational start site. The CC genotype was associated with Graves’ disease and gave a relative risk of 1.6. Previous studies using a mixed population of patients had not supported such a linkage, but in a Caucasian population, this association and linkage were shown(150). The frequency of C/C genotype of CD40 was increased in GD compared to controls, but the difference was not significant (60.5% versus 55.8%, p = 0.062, odds ratio [OR] = 1.21, 95% confidence interval [CI]: 0.96-1.53). In a meta-analysis with the data from previous studies, the combined OR for the C/C genotype as a risk factor for GD was 1.22 (95% CI: 1.08-1.38, p = 0.001). There was no interaction between CD40 genotypes and other GD susceptibility alleles. No significant genotype-phenotype associations were found. The CD40 C-T polymorphism appears to have a modest effect on genetic susceptibility to sporadic GD(151).
A promoter polymorphism of the CYP27B1 gene has been associated with Graves' Disease and other autoimmune diseases (152). Interestingly, this gene catalyzes the conversion of 25-OH-D to 1,25-OH-D, the active metabolite of D. A possible relation of Vit D receptor to Graves' was noted above. A B cell specific gene ("ZFAT") has been linked to autoimmune thyroid disease, though not specifically to Graves (153). IL-13 gene polymorphisms were studied in Japanese GD patients and healthy control subjects without antithyroid autoantibodies or a family history of autoimmune disorders. A C/T polymorphism at position -1112 of the promoter region was measured using the direct sequencing method, and an Arg-Gln (G2044A) polymorphism in exon 4 was examined using the PCR-restriction fragment length polymorphism method. IL-13 gene polymorphisms are associated with GD susceptibility in Japan(154).
The lymphoid tyrosine phosphatase, encoded by the protein tyrosine phosphatase-22 (PTPN22) gene, is a powerful inhibitor of T cell activation. Recently, a single-nucleotide polymorphism (SNP), encoding a functional arginine to tryptophan residue change at PTPN22 codon 620 in Caucasians has been shown to be associated with GD and other autoimmune diseases. This variation inhibits function of the gene, which is normally to down-modulate signaling via binding to Csk and phosphorylation of Lck. Using a polymerase chain reaction (PCR)-restriction fragment (XcmI) assay to examine genotypes at the codon 620 polymorphism in 334 unrelated patients with AITD and 179 controls, none of the patients with AITD and controls had the tryptophan allele. Of interest, knockout mice deficient in this gene do not develop signs of autoimmunity, suggesting it may not be important in etiology of AITD (155).
The +869T/C polymorphism in the TGF-beta1 gene has been  associated with the severity and intractability of both Graves’ disease and thyroiditis(156). CD25 has been related to Graves’ disease, and IL-23R to ophthalmopathy(157). A significant association of the Interferon Inducible Helicase,  Ala946Thr, IFIH1 polymorphism to organ-specific autoimmune diseases including Graves' disease.was reported by Sutherland et al (158). IL-1alpha and IL-1beta polymorphisma are associated with Graves’on Asian populations (159).   Gene associations with GD or AITD, and a long list of othergenes-TNFAIP3, FoxP3, TBX21, HLX, BTNL2, Notch4, and CXCR4, has been reported.

Although linkage analysis has often been considered to be superior to gene association studies for determining genetic effects in autoimmune diseases, in fact linkage analysis may be limited in defining such loci, and large-scale association studies may prove to be more useful in identifying genetic susceptibility factors for AITD. A genome-wide screen was performed on affected relative pairs with autoimmune thyroid disease. 1119 Caucasian relative pairs affected with autoimmune thyroid disease (GD or AIH) were recruited into the study. The study aimed to identify regions of genetic linkage to AITD. Three regions of suggestive linkage were obtained on chromosomes 18p11 (maximum LOD score 2.5), 2q36 (maximum LOD score 2.2) and 11p15 (maximum LOD score 2.0). No linkage to HLA was found. The absence of significant evidence of linkage at any one locus in such a large dataset argues that genetic susceptibility to AITD reflects a number of loci each with a modest effect (78.17).

The final result of this kind of research is not clear at present. The most obvious conclusion is that there are several -- probably very many -- genes which augment the possibility of developing immunity to thyroid gland protein components. Thus inheritance is polygenic. Rather than inheriting one gene which, in a dominant fashion, induces Graves' disease, individuals inherit many different genes which are related to the development of thyroid autoimmunity. If a sufficient load of the positively acting genes is inherited, they support the development of Graves' disease or other AITD, especially if other factors are present such as injury to the thyroid. A dramatic illustration of the genetic influence is provided in a recent report of "adoptive" hyperthyroidism following allogenic stem cell transplant from an HLA-identical twin with Graves’.[161]

Gender

Perhaps the clearest association with autoimmune disease is being a member of the female sex, which carries a 10 - 20-fold risk compared to the male sex. Despite this obvious association, the mechanism has remained obscure. The association carries through not only for autoimmune thyroid disease but also for the development of multinodular goiter, and even differentiated thyroid carcinoma, but not undifferentiated thyroid carcinoma. Thus female gender may endow a generally greater reactivity of the thyroid gland, or may subject it to greater stress in some manner. It has been suggested that there may be specific receptors on the promoter for DR genes, which makes them responsive to the estrogen receptor. In a Polish population the ESR2 A allele is associated with GD with a strength comparable to polymorphisms of PTPN22 and CTLA4 CT60 loci (OR approximately 1.7). The association with ESR2 is found in both sexes and may be particularly strong among the DRB1*03-negative individuals(162)

Other Suggested Causative Factors

A variety of other ideas have been presented as the "cause of Graves' disease", but remain unlikely or unproven. ( Table 10-3) Mutation of T or B cells to produce a specific reactive clone has been suggested [163]. Of course somatic mutation is a known part of the development of B cell clones, and specific mutations of B cells or T cells can produce tumors rather than a disease producing autoimmunity.

Regulatory T cell abnormalities

Every immune response involves development of a set of effector T cells and at the same time a set of regulatory T cells designed to temper the response. Current research gives the regulatory cells a prominent role in controlling anti-self immunity, as seen in myasthenia, multiple sclerosis, thyroid autoimmunity, and Type 1 Diabetes. A common assumption has been that numbers or function of Tregs would be deficient in human autoimmunity, since mouse knock-out models and human mutations leading to deficient expression of FoxP3 (and CTLA-4) lead to broad and sometimes fatal autoimmunity(164).
An important class of Tregs is characterized by expression of CD4, CD25 (IL-2Rα subunit), FoxP3, GITR (glucocorticoid inducible TNF receptor), CTLA-4 (cytotoxic T lymphocyte associated antigen), GADD45 alpha and beta (growth arrest and damage inducible proteins), and a low level CD127 (IL-7-R subunit alpha chain) (165). The FoxP3 transcription factor is one hallmark (but not always reliable) for Treg suppressor function (166,167). CTLA-4 is a negative regulator of T cell proliferation. Triggering of the Stat signal pathway by IL-2 and IL-7 is essential for survival and proliferation of Tregs.
Tregs are comprised of several different cell types. “Natural” Tregs (nTreg) develop in utero within the thymus, recognize self-antigen, and are CD4+CD25+GITR+FoxP3+. They are IL-2 dependent and commonly anergic in vitro in the presence of antigen(168). Inducible Tregs (iTregs) develop in the periphery from CD 25lo cells (169). TGFbeta can promote the development of CD4+CD25- T cells into CD4+CD25+CTLA-4+, GITR+,FoxP3+ cells, unless exposure to IL-6 re-directs their development into Th17 effector T cells expressing RORgammaT (170) (Fig. 1.3). Tregs have been shown to function by cell-cell contact or in some systems by secretion of TGFbeta and/or IL10 and may directly suppress effector T cell reactivity, or act via induction of other Tregs (171). Natural Tregs also induce autoantigen-specific adaptive Tregs (172,173).Several other cell types such as CD8+ cells and CD69L+ cells (173) also have been shown to function as regulatory T cells, and their importance in Graves’ disease is unknown. The ability to induce non-antigen-specific bystander suppression is a hallmark of Tregs.

Tolerance can develop in the periphery when T cells are converted to an adaptive (aTreg) (or inducible (iTreg) Treg phenotype upon activation via their TCR in the presence of IL-10 and TGF-b. The role of these iTreg cells may be to dampen over-emphatic inflammatory immune responses. iTregs can suppress T helper responses, B cell responses (antibody) and CTL responses. iTreg induction also is associated with sustained tolerance (to autologous proteins) and probably requires the existence of Treg cells with the same antigen-specificity as the self-reactive (effector) T cells. Unfortunately the expression of FoxP3, while characteristic of regulatory T cells, is neither unique nor stable. CD4+FoxP3+ cells do not necessarily display a suppressive function, and more  problematically, can be converted in the right milieu, into cells espressing Th17 with an effector function

The role of regulatory cells during development and progression of autoimmune disease has been widely studied.  For example, the function of Treg cells appears to be impaired during development of diabetes in NOD mice (174,175). During development of EAE in mice, Tregs fail to suppress the immune response. (176). CD4+CD25High Tregs from patients with active SLE failed to suppress function of CD4+ effector cells in vitro, whereas Tregs from patients with inactive disease were effective(177). CD4+CD25+FoxP3+ cells are reported to be present in normal numbers but to have deficient suppressor function in patients with ITP (178). Tregs are reported to be decreased in number in myasthenia gravis, while CD8+CD122+ Tregs are normal (179). Many other studies on this topic cannot all be cited.

Several groups have investigated Tregs in AITD (Autoimmune thyroid disease) with conflicting results. Nakano et al. found that the proportion of Tregs among intra-thyroidal lymphocytes was lower than among PBMC in patients with GD and thyroiditis and that the Tregs present were apoptotic (180). Marazuela et al. (181) found an increased percent of CD4+ T cells expressing GITR, FoxP3, Il-10, TGF-beta and CD69 among PBMC from patients with autoimmune thyroiditis and that similar cells infiltrated Hashimoto’s thyroiditis tissue. The suppressive function of blood Tregs was defective. Gangi et al. found that administration of GM-CSF  induced development of CD4+CD25+ Tregs that suppressed immunity to TG in mice (182). Molteni et al. reported that CD8+ T cells suppressed TSH-R specific CD4+ T cell clones (183). Watanabe et al. reported that relative proportions of CD25+ cells among CD8+ cells, and CD4+ cells were directly related to severity of Hashimoto’s thyroiditis (184). Vaidya et al. reported higher numbers of naïve activated T cells (CD4+ or CD45RA+CD4+) and lower memory T cells (CD45RO+ CD4+) in patients with GO as compared to controls(185). Glick et al found frequency of Tregs normal in  20  HT and GD children but impaired Treg suppresion of Teffectors(186).  Klatka et al found lower numbers of Tregs and increased numbers of Th17 cells in adolescents with GD, returning towardnormalwith treatment (187). Our own data indicate  that Tregs are normal as a percentage of cells and cell numbers in treated patients and those with active GD (188). In contrast, Mao et al (189) report that circulating FoxP3+Tregs are diminished in untreated GD, and that their function is further reduced by plasmacytoid  dendritic cell secretion of IFNalpha and elevated thyroid hormones.
While there is much evidence for abnormalities in Treg number and/or function in GD, there remains some uncertainty.  On the otherhand,  it is a reasonable assumption that Tregs are at some time, or in some way, relatively inadequate, or patients would not develop GD(189-193).

It is probable that a relative deficiency of such cells explains the appearance of GD during immune system recovery following medical treatment of patients with advance HIV disease (78.3) and after therapy with the lymphocyte depleting antibody CAMPATH.

Figure 1.3. After activation, CD4+ T cells may enter several different pathways depending on APC co-stimulatory factors and cytokine milieu. TGF-beta directs T cells to become Tregs, unless IL-6 is present, in which case the cells may be acted on by IL-21 and IL-23, leading to expression of RORgammaT,and STAT 3, and becoming Th17 or IL-17a effector and inflammatory cells. FoxP3+ Tregs can be converted into Th17 cells. Pathways to Th1 and Th2 cells, and secretory products, are also shown.

Fetal cell microchimerism

Intrathyroidal fetal cell microchimerism has been suggested as a possible etiologic agent in autoimmunity. During pregnancy, fetal and maternal cells are transferred between mother and fetus. It has been shown that fetal cells from male infants can persist in the maternal circulation for up to 20 years. Male fetal origin cells were studied in human thyroids by identifying the male specific region of the SRY region of the Y chromosome, and were detected in 6 of 7 frozen thyroid tissue specimens from patients with Graves’ disease, and one of four with thyroid nodules. Fetal male cells are possible candidates for modulating autoimmune thyroid disease, since they might either induce an immune response, or develop a sort of graft-versus-host immune response to the mother (194).

Table 3. Other Factors Suggested in the Etiology of Graves' Disease

Psychic Trauma

•  Sympathetic "Overactivity"

•  Weight Loss

•  Iodine

•  TSH

•  Female gender

Development of expression of Class I or Class II MHC

Development of expression of Class I or Class II MHC molecules on the thyroid epithelial cell was suggested as a factor in the causation of Graves' disease by Bottazzo et al [195]. It is now apparent that exposure of thyroid epithelial cells to Interferon, presumably elaborated by infiltrating lymphocytes or other immune cells, can lead to the expression of Class II molecules on the thyroid cell surface [196]. Expression of these molecules does allow the thyroid epithelial cell to function as a weak antigen presenting cell [197]. Class II expression is secondary to the effect of an autoimmune lymphocyte attack and is induced by Interferon [198]. Culture of human thyroid cells from patients with Graves' disease in vitro shows that Class II expression disappears [199], as it does when the cells are transplanted into nude mice [200].

It is also possible that the Class II expression is a defensive response [201] Antigen presentation by Class II MHC expressed on a thyroid cell,to a T cell, in the absence of a second signal, could lead to anergy of the attacking T cell. Possibly Class II expression is secondary but may play a role in continuing or strengthening the autoimmune reactivity to thyroid antigens. Antithyroid drugs may have an immunosuppressive effect on autoimmune thyroid disease through inhibition of HLA-DR expression on thyrocytes. Follicular cells of patients with overt thyrotoxicosis express HLA-DR, while those in remission, or those under medication with antithyroid drugs, did not. Recently it was reported that transgenic mice expressing MHC Class II on their thyroid cells do not develop autoimmunity. This is a strong argument indicating that Class II MHC expression on Graves thyroid cells is secondary and not a causative event.

Stress

Psychic trauma or psychic stress has long been considered to be a possible etiology of Graves' disease. In Perry's original report, a crippled woman, who was injured when her nanny allowed her wheelchair to role down a flight of stairs, had rapid onset of thyrotoxicosis. This report, in 1820, has been followed by many more studies, some of which support the idea. The incidence of Graves' disease increased in Denmark during World War II [202], but did not in Ireland during the sustained civil war in that country during the period of 1980 through 1990. A recent study in Yugoslavia indicated that patients with Graves' disease had suffered on average more stressful episodes than control subjects, but previous similar studies have failed to show this relationship [203-205].  A recent article found increased numbers of stressful life events in patients with Graves’ disease prior to onset of the disease, compared to patients in a toxic nodular goiter group who had a similar number as control patients. (206). Stress induces a variety of physiologic responses including anxiety, tachycardia, restlessness, etc., which are not unlike symptoms of Graves' disease. Its role remains enigmatic in causation of Graves' disease to this date. A mechanistic route from stress to the development of Graves' disease is not obvious. Theoretically, stress might cause activation of the adrenal cortex or the sympathetic nervous system. Hypercortisolism would tend to suppress autoimmunity. Heightened sympathetic nervous system activity might theoretically cause stimulation of thyroid secretion, as has been shown in experimental animals [207. Other specific stressors have been reported. Aggressive weight loss programs have been reported to induce Graves' disease. Administration of thyroid hormone, sometimes given for induction of weight loss, also has been followed by mini-epidemics of Graves' disease [208].

Excess Iodide

Iodide itself has been thought to induce Graves' disease, thus leading to the term "Jod Basedow". This syndrome refers to the occurrence of thyrotoxicosis following supplementation of iodide in medicinals or by salt iodinization. Excess iodide clearly does induce hyperthyroidism in patients with multinodular goiter [209,210]. Presumably autonomous nodules in the goiter are unable to produce an excess of hormone when their synthesis is limited by iodide, but when this iodide supply is augmented many-fold, the nodules can process it to produce an excess of hormone. The best defined epidemic of iodide-induced thyrotoxicosis occurred in Tasmania after the government introduced iodization of salt, and was clearly associated with multinodular goiters rather than typical Graves' disease [211,212].

Possibly increased iodide intake can actually augment thyroid autoimmunity through other mechanisms. For example, increased iodide intake has been correlated with an increase in incidence of AITD [213This could in theory work by augmenting iodination of TG, and heavily iodinated TG is more immunogenic in animals than is poorly iodinated TG. Also, under special circumstances excess iodide can induce thyroid cell necrosis, and this might liberate antigens. Adding KI to the diet of the thyroiditis-prone BB strain rat and to NOD mice increases the severity of thyroiditis [214,215].

Whether an excess of iodide can induce true Graves' disease and autoimmunity remains unknown. In fact the addition of 2 - 6 mg per day of iodide to the intake of most patients with Graves' disease, raising plasma iodide levels above 5ug/dl, causes a dramatic reduction in hormone release by the "Wolff-Chaikoff" effect, which is an inhibition of hormone synthesis and of hormone release [216-219]. Iodide is one of the most rapid acting agents in suppressing thyrotoxicosis. While it has this effect in most individuals with Graves' disease, its action tends to be partial or transient, and thus is not relied upon as an effective antithyroid agent. Occasional contemporary reports attest to the ability of iodine treatment to induce remission of Graves” disease in a significant proportion of children over several years. A recent study suggests that the ability of iodide to suppress Graves' disease may be because iodide down-regulates MHC Class I and II expression on thyroid cells [220].

Smoking has been related to Graves' Disease, and more specifically to a greater propensity to develop ophthalmopathy, or to have worsening of the condition. Salvi et al also studied cytokines in patients with Graves’ disease and found serum IL-6 concentrations higher. Interestingly, smoking, which is associated with an adverse effect on Graves’ ophthalmopathy, appeared to have no interaction with the serum lymphokines [221].

Other older ideas on the cause of Graves' disease included a role for TSH, or some fragment of TSH [108], or the HCG molecule. None of these is thought to be involved in thyroid autoimmunity according to current formulations, although excess TSH production by a pituitary adenoma is an established cause of thyrotoxicosis [223, 224], and TSH produces the thyrotoxicosis seen in "Pituitary Resistance to Thyroid Hormone". Vassart and co-workers [225] have recently recognized the cause of non-autoimmune Hereditary Familial Hyperthyroidism inherited in an autosomal dominant manner. In two sibships, activating germline mutations were found in the transmembrane segments of the TSH-R. The mutations cause persistent basal hyperfunction of the receptor and early onset of thyrotoxicosis. Other causes of thyrotoxicosis (but not Graves' Disease) are described briefly in Chapter 11 and more fully in Chapter 13. Numerous other theories regarding the cause of Graves' disease have been proposed in the past. For a critical review of earlier speculations and a superb bibliography, the reader is referred to the monograph by Iversen [226].

THYROID GLAND FUNCTION IN GRAVES’ DISEASE

While the ultimate cause(s) remain uncertain, the patho-physiologic mechanisms are more clear. The thyroid gland is functioning at an accelerated rate. Cell membrane adenyl cyclase activity in tissue from Graves' patients is higher than in normal thyroids, and the increment coincides with TSAb in the serum [227], which acts on TSH receptors to cause the response. Clearance of plasma iodide by the gland is increased from the normal rate of 10-20 ml/min to 40 to several hundred ml per minute. We have estimated iodide clearance in one patient to exceed 2 liters/min. For this reason, the percentage of a tracer dose of 131I (the radioactive iodine uptake or RAIU) found in the gland at 24 hours is characteristically elevated. Thyroid peroxidase activity is increased. Because of rapid secretion, the period of retention of iodine within the thyroid is reduced, causing the characteristic drop in RAIU between 12 and 24 hours after administration of a tracer. The total quantity of iodine in the gland is variable. In previous years it tended to be reduced, but now it is often elevated because of increasing amounts of iodine in our diet. The volume of the gland is characteristically but not invariably increased. Thyroid hormones, TG, small amounts of an iodinated albumin-like protein [219,228], and iodotyrosines [229] are released into the blood at increased rates. The latter two components are normally either not secreted or are released in minute amounts.

Many studies with radioiodine have confirmed the accelerated physiologic activity of the thyroid in Graves' disease. Thus, labeled hormones appear as plasma Protein Bound 131I more rapidly and reach higher levels than in normal persons after administration of 131I. The rate of turnover of plasma hormones is also increased. Accelerated degradation is probably secondary to hypermetabolism and not a primary event [230], although it has been reported that accelerated T4 turnover persists after therapy for thyrotoxicosis [231].

With the general hyperactivity of the thyroid, excess TG is released and serum TG levels are elevated in active Graves' disease. After therapy the levels tend to fall, and normalization during antithyroid therapy is an excellent predictor of remission [232]. This excessive release of TG leads to formation of circulating immune complexes with anti-TG antibodies, and can lower the titer of these antibodies in the serum.

An important abnormality in thyroid function during Graves' disease is that the uptake of 131I by the thyroid is not suppressed by administration of exogenous T4 or T3 ( 233, 234). This fact holds true even if large amounts of hormone are given. Indeed, administration of T3 may cause, on average, a slight increase in the 24-hour radioiodine uptake. This abnormal response to the administration of thyroid hormone occurs in spite of responsiveness of the thyroid to administered exogenous TSH, as measured by augmented release of thyroid hormone. It may persist after thyrotoxicosis has been ameliorated by surgery, but typically suppressibility returns in time after treatment. Non-suppressibility is observed so regularly that it was used for many years as a criterion for diagnosis in doubtful cases. Non-suppressibility is caused by stimulation of the thyroid by TSAb, and independence of feedback control via TSH. There is a general, but not complete, correlation between T3 nonsuppressibility and positive assays for TSAb [235]. Even long after apparent remission of Graves' disease, some patients show some resistance to T3 suppression or TSH stimulation [236]. Also, some euthyroid relatives of Graves' disease patients show these abnormalities in the absence of overt thyroid disease.

The pituitary does not respond to TRH in the thyrotoxic state, either in hyperthyroid Graves' disease or when thyroid hormone is administered. This is because the function of the hypothalamic TRH neurons are down-regulated by excess thyroid hormone levels. Some patients with "euthyroid Graves' disease" respond to TRH and others do not; the responses do not correlate with the results of T3 suppression tests [237, 238]. For the diagnosis of Graves' disease, suppression of serum TSH is currently the most useful criterion, as it is more convenient than the TRH test and preferable to the T3 suppression test in patients with heart disease. Test results are not uniformly abnormal in patients with euthyroid Graves' disease.

IODINE SUPPRESSION OF THE GRAVES’ THYROID

Iodide affects the metabolism of the diffusely hyperplastic thyrotoxic gland in a way radically different from its action on the normal gland. Years ago, Plummer demonstrated that Graves' disease can be temporarily or permanently controlled by the administration of iodide [239]. The amount needed is 6 mg/day or more. Administration of large doses of iodide to laboratory animals causes a temporary inhibition of iodide organification, the Wolff-Chaikoff block. High intrathyroidal iodide concentration is the crucial factor inducing this response [240]. The same phenomenon occurs in humans, and thyrotoxic patients are especially sensitive to this effect. The thyroid uptake of 131I is acutely depressed in thyrotoxic patients by administration of 2 mg potassium iodide, whereas more than 5 mg is needed to depress uptake in normal subjects. Concentrations of serum iodide above 5 µg/dl block binding in the thyrotoxic gland [216-219].

The biochemical mechanism of the Wolff-Chaikoff block is not clear. Iodide does not prevent TSH or TSAb binding to the TSH membrane receptor, but does inhibit both TSH-stimulated adenyl cyclase production of cAMP, and cAMP actions. Since iodide inhibition of cAMP production and action is blocked by methimazole, it is hypothesized that an oxidized iodide intermediate is involved. Alternatively, the block of iodination may be caused by depression of H2O2 generation. Whether the inhibition of iodide transport and binding relate to the recognized changes in cAMP formation is not known (see also Chapter 5).

In animals the block of iodide binding is transient; during continuous iodide administration, binding recommences. This escape also occurs in most normal humans. Few individuals who take large doses of iodine continuously ever develop myxedema. Adaptation to excess iodine in animals involves a reduction of iodide transport into the thyroid, a lowering of intrathyroidal iodide content, and escape from the Wolff-Chaikoff block. This adaptation occurs independently of TSH action. Possibly because the Graves' gland is hyperactive under continued stimulation by TSAb, it may remain blocked by administered iodide, and hormone production may remain suppressed. (Actually in about 1/3 of patients the gland is only partially blocked, and in another third escape occurs after a few weeks.) A similar sensitivity to inhibition by iodide occurs in Hashimoto's thyroiditis, hyperfunctional adenomas, and possibly the normal gland when stimulated by exogenous TSH [241]. Myxedema can often be induced by administration of iodide to patients who have had a partial thyroidectomy for Graves' disease [242-243]. Thus, there is an inherent susceptibility of these glands to the action of iodine. Sensitivity of the gland in Graves' disease to iodide is also demonstrated with the iodide-perchlorate test [242]. In this test, it is seen that the dose of iodide required to block organification in Graves' disease is much smaller than that in the normal subject. The inhibition of binding by iodide is revealed by administration of perchlorate, which discharges the accumulated 131I present in the gland as free iodide.

Coincident with the block in uptake, iodide also causes a marked reduction in the release of previously formed hormone from the thyrotoxic gland. This phenomenon has been repeatedly observed and helps to explain the beneficial therapeutic effect of iodide in Graves' disease, as originally recognized by Plummer. Iodide administration blocks release of hormone from the gland but does not completely prevent hormone synthesis, for under these circumstances the gland gradually accumulates an increased store of organic iodine. Ochi et al [244] have shown that chronic administration of iodide in Graves' disease blocks the stimulating effect on hormone release of both TSH and TSAb. It is this block of release, rather than a block of hormone synthesis, that is responsible for the dramatic rapid beneficial therapeutic effects of iodide administration [245]. The block of hormone release that occurs in the thyroid of Graves' disease can be observed, although not uniformly, in the normal gland and in the normal gland made hyperactive by repeated administration of exogenous TSH. Iodide also inhibits the release of hormone from autonomous hyperfunctioning adenomas, presumably in the absence of endogenous TSH. This observation also indicates that iodide block is by a direct action on the thyroid gland.

Thus, the Graves' gland appears to be unusually sensitive to small amounts of iodide, as manifested by (1) a block of iodide uptake and binding and (2) a block of hormone release. Perhaps these are two parts of the same fundamental process. Sensitivity to iodide may be related to the TSAb dependent, hyperactive, iodide-concentrating mechanism of the Graves' gland.

A further abnormality in intrathyroid iodine metabolism is that the toxic gland continuously spills into the circulation large amounts of nonthyroxine iodide, in addition to hormone [246]. The iodide may be a product of the deiodination of iodotyrosines released from TG during its hydrolysis.

INCIDENCE AND DISTRIBUTION OF GRAVES' DISEASE

The incidence of Graves' disease in Olmstead County, MN was found to be 30 cases per 100,000 annually [247]. A thorough examination of an English town by Tunbridge and associates found an incidence of 100 - 200 cases per 100,000 per year, significantly higher than the previous estimates [248]. In this report, it was also found that 2.7% of women and .23% of men had either current Graves' disease or a history of Graves' disease. This survey also noted that goiter was present in 15% of women, antithyroid antibodies in 10.3% of women, and that hypothyroidism was about two-thirds as common as Graves' disease. A recent update in this area showed a continuing incidence of 80 cases/100,000 women/year [249]. The prevalence of hyperthyroidism was found in the “HANES” study to be 0.5% at the clinical level and 0.7% at the sub-clinical level (250). Data attest to a lifelong incidence of autoimmune thyroid disease of > 6%, comprised roughly equally by Graves' disease, Hashimoto's thyroiditis and idiopathic hypothyroidism.

The distribution of Graves' disease around the globe, so far as data is available, appears to be relatively equal, affecting all countries and races.

Graves' disease is most typically a disease of adult women in the age group between 30 and 60, and has an incidence roughly eight times greater in women than in men [248-249]. Aside from the infrequent occurrence of postnatal thyrotoxicosis due to maternal antibodies, the incidence of spontaneous Graves' disease in children before the age of ten is most unusual, but the incidence climbs with each decade until about age 60 [248-249, 250]. The greater incidence in women is typical of most thyroid diseases including multinodular goiter and differentiated thyroid carcinoma, and the mechanism for this association is unknown. One possibility is that female reproductive activity somehow stresses the thyroid. Another possibility is that the promoter for certain genes such as Class II HLA molecules may have estrogen receptor response elements and thus be activated more easily in women. The well known familial distribution of Graves' disease, recognized by all clinicians caring for patients, is thought to be explained by inheritance of specific genes, as detailed previously.

PATHOLOGY

It should be noted that only the abnormalities of the thyroid, orbital contents, lymphatic system, and skin can be considered specific for Graves' disease; the other lesions probably could be caused by thyrotoxicosis of any cause.

The Thyroid

It is known from observations made before the introduction of iodide or antithyroid drugs that the essential lesion of Graves' disease is parenchymatous hypertrophy and hyperplasia (Figure 10-2). The central features are increased height of the epithelium from cuboidal to columnar; and redundancy of the follicular wall, giving on section the picture of papillary infoldings, cytologic evidence of increased activity, hypertrophy of the Golgi apparatus, increased number of mitochondria, and increased vacuolization of colloid. There is probably nothing specific about the hyperplasia. Any stimulus that calls for sustained hyperfunction produces this picture, such as antithyroid drugs. There is also a characteristic lymphocyte and plasma cell infiltrate. This infiltrate may be mild and diffuse throughout the gland, but more typically occurs as aggregates of mononuclear cells and even lymphoid germinal centers, referred to as focal thyroiditis. Occasionally the histologic pattern completely overlaps that of Hashimoto's thyroidits.

Figure 2. Extreme thyroid hyperplasia in Graves' Disease, with tall cells, small follicles, scant and "scalloped" colloid. Figure kindly provided by Dr. Francis Straus.

Fine structure examination discloses a rather widely varying size and shape of the follicles, with columnar cells and reduced homogeneous colloid [250, 251].The basement membrane is well demarcated and is 400 - 1,000 A thick. Between the follicles is a large array of capillaries, together with lymphocytes and fibrocytes. The apical end of the follicular cell often bulges into the lumen, and cuplike villi extend into the lumen. Vesicles and free ribosomes may be found in these villi. These microvilli vary in size and shape; some may enclose colloid. The nucleus is near the basal part of the cell. The mitochondria are numerous; they are mostly large, elongated structures, and some are branched. The endoplasmic reticulum is usually well developed. Ribosomes occur in great numbers. The Golgi apparatus is well developed. Vesicles are present in abundance, but vary in size and number from cell to cell. Multivesicular bodies are occasionally found near the Golgi apparatus. Droplets, globules, and dense bodies appear. Phagolysosomes are common. All these changes are quite like those of the normal thyroid chronically stimulated by TSH. 

There are some data on the thyroid in persons who have recovered from Graves' disease [252]. Autopsy material from seven patients who had recovered from exophthalmic goiter and died later of other causes showed complete regression of the hyperplastic changes.

EXTRATHYROIDAL CHANGES

The ophthalmologic problem and pretibial myxedema, which are unique to Graves' disease, are described in chapter on Complications of Graves’ disease.

Abnormalities in striated muscle may be a part of Graves' disease [252-254 ]. Decades ago Askanazy and Rutishauser [255] studied four patients with hyperthyroidism on whom autopsies were performed. They found a diffuse process in all striated muscles, including the extrinsic muscles of the eyeball, consisting of degenerative atrophy of muscle cells, fatty infiltration, loss of striation and uniform appearance, vacuolization, and proliferation or degeneration of nuclei. Cardiac and smooth muscle were not involved in this process. Not all muscle groups were equally involved, nor were the same muscles involved in different patients.

Dudgeon and Urquhart [256], in studies of the muscles in nine postmortem cases of Graves' disease, found in various skeletal muscles interstitial myositis characterized by plasma cells, tissue macrophages, and atrophy of fibers. Ocular muscles were more affected and cardiac muscle less affected than skeletal muscle. The lesions were spotty and were observed in only a small fraction of the sections. On the other hand, Naffziger [257] examined biopsy muscle specimens from other parts of the body in patients with the ophthalmopathy of Graves' disease and found no abnormalities. The fat content of skeletal muscle may be increased, just as it is in the extraocular muscles [258]. Myocardial degenerative lesions have been reported in thyrotoxicosis, with foci of cell necrosis, mononuclear infiltrates, and mucopolysaccharide deposits similar to those described in extraocular and skeletal muscles [258],and severe damage has been found in patients dying of thyrotoxicosis [146]. (Figure 10-3).

Figure 3. Myocardial tissue from a patient that died of cardiac failure with extreme thyrotoxicosis, showing degeneration of myocardial cells, mononuclear and polymorphonuclear infiltration, and edema.

The extraocular muscle lesions are probably specific for Graves' disease, whereas the remainder of the abnormalities may reflect the action of excess hormone. 

The anterior pituitary demonstrates, not surprisingly, a dramatic decrease in identifiable thyrotropin containing cells in patients dying in thyroid storm. This loss is found entirely reversed in patients who come to autopsy after treatment to euthyroidism [260-261].

Lesions specific for Graves' disease do not appear in the parathyroids, gonads, or pancreas.

Studies based on autopsies of patients with Graves' disease made years ago demonstrated focal and even diffuse liver cell necrosis [262], atrophy, and cirrhosis, including a kind of peripheral fibrosis that was believed to be peculiar to this disease -- cirrhosis basedowiana. In more contemporary series of liver biopsy specimens obtained from thyrotoxic persons, the deviations from normal were minimal [263, 264]. Some decrease in glycogen and increase in fat, and some round cell infiltrates were noted. The differences among these studies are at least superficially explicable on the basis of lesser severity and duration of the disease in those patients studied during life.

Hyperplastic changes may be found in the spleen, thymus, and lymph nodes in Graves' disease. The thymus occasionally presents as an anterior mediastinal mass [265] and has been inadvertently resected. Persistence or enlargement of the thymus was once believed to be significant in Graves' disease, and early in the century thymectomies were performed for its treatment, with apparent benefit. The TSH-R is expressed in thymic tissue, suggesting that it might be the target of auto-immunity inducing hyperplasia.

Prolonged hyperthyroidism is known to produce the histologic picture of osteoporosis[266], but osteitis fibrosa also occurs [267]. Histomorphometric studies show clear evidence of excess bone formation and resorption. The high degree of exchangeability of calcium in the bones of patients with thyrotoxicosis and the high rate of loss of calcium in the urine are discussed later in this chapter. Serum 1,25-dihydroxychole-calciferol is decreased, probably in response to increased bone turnover [268].

Cytologic investigations of mitochondria using the electron microscope have revealed anatomic lesions not visible by the ordinary light microscope. Schulz et al [269] reported that the mitochondria from tissues of T4-treated animals appeared to be swollen.

DEVELOPMENT OF THE CLINICAL PICTURE AND THE COURSE OF THE DISEASE

Graves' disease displays an array of possible clinical patterns extending from that of goiter and thyrotoxicosis, but without ophthalmopathy, to that of ophthalmopathy without goiter or thyrotoxicosis.

In classic exophthalmic goiter, or Graves' disease, the most common onset is the simultaneous and gradual development, over a period of weeks or months, of the symptoms of thyrotoxicosis, enlargement of the thyroid, and prominence or related abnormality of the eyes (Figure 10-4). It is quite possible for classic Graves' disease to develop in a patient with preceding (probably unrelated) nontoxic goiter. This is common in a goitrous country. Often the onset of symptoms is so gradual that it is difficult or impossible for the patient or physician to fix its date. The more abrupt onset may sometimes be sufficiently rapid to justify the term fulminating. The picture of classic full- blown exophthalmic goiter has appeared in a person apparently previously well in as short a period as two to five days. Rare patients have first developed hypothyroidism and later thyrotoxicosis [54], probably because the initial development of TBAb is followed by some natural immune modulation and development of stimulatory antibodies. Continuous significantly elevated titers of anti-TSH-R antibodies  in the months after diagnosis are positively correlated with serious ophthalmopathy (55).

Figure 4. Classic severe Graves' ophthalmopathy demonstrating a widened palpebral fissure, periorbital edema, proptosis, chemosis, and conjunctival injection.

In many patients, the symptoms of Graves' disease are first noted after some emotional trauma. These associations are certainly of importance in understanding the patient's backgound, but, as noted above, whether or not they bear a causal relationship to the development of Graves' disease remains conjectural. 

As noted in Chapter 14,Graves' disease is frequently partially or totally suppressed during pregnancy, and initial or recurrent manifestations can occur in the postpartum period. (Figure 10-10) Sometimes the "painless thyroiditis" characteristic of this period co-exists and masks the development of Graves' disease .

Weight reduction, as mentioned above, has also constituted an activating episode in Graves' disease.

At present, the natural history of the thyrotoxic process is usually altered by definitive therapy. Before the general availability of good treatment, hyperthyroidism tended to progress through periods of exacerbation and remission. In perhaps a quarter of the patients, especially those with a mild form of the disease, the process was self-limited to one year or more, as the patients returned spontaneously to a euthyroid state.

M.S., 27-Year-Old-Man: Thyrotoxicosis with Spontaneous Remission

This young physician developed tachycardia, hyperkinesis, decreased heat tolerance, slight tremor, and weight loss over three or four months. On examination, blood pressure (BP) was 150/50, pulse rate 86, and the skin was sweaty. There was a fine tremor. The eyes were entirely normal. There was a grade 1 precordial systolic murmur. The thyroid was about twice the normal size, diffusely enlarged, and firm. There were several cervical lymph nodes bilaterally. PBI was 11(nl 4--8ug/dl), and the rT3U level was elevated. RAIU was 57% and BMR-10. All tests were repeated once, and the results all remained as indicated.

The patient was given 100 mg PTU three times daily and was maintained on this program for 18 months. During this time, the T4 level was maintained in the range of 7.3 ug/dl and the FTI in the range of 6 (nl 4-10); the white cell count remained normal. The TGHA titer was 1/320, and there was a borderline positive TSAb bioassay response. During the course of therapy, the 20-minute technetium uptake test was repeatedly measured while the patient received both antithyroid drugs and suppressive doses of T3; suppressibility of the thyroid gradually fell to the normal range. Eighteen months after the initiation of therapy, the patient developed an acute gastroenteritis and was briefly hospitalized. At this time, because of the possible association of PTU with gastric irritation, the medication was discontinued.

He subsequently remained well for three months, but then developed symptoms of mild hyperthyroidism. The thyroid was again found to be two to three times the normal size, the T4 level to be 10.3 µg/dl, and the FTI to be 11.9. Since the symptoms were mild, it was elected to observe events without therapy for a period. Initially, the symptoms, signs, and laboratory test results remained abnormal, but over several months the mild tachycardia, increased sweating, and increased nervousness gradually dissipated. Six months later, the T4 level was 6.7 µg/dl and the FTI 8. The TSAb bioassay result remained positive. No further treatment was given, and the patient has remained entirely well with a moderate thyroid enlargement, normal thyroid function test results, and no symptoms over the subsequent 30 years.

In one of the few documented reports of untreated thyrotoxicosis, White [270] found that of 12 patients, 7 died in an average of three and a half years and the remainder lived on without therapy. From a large series, Sattler estimated that in the past mortality was up to 11% [271].  Fortunately, death due to hyperthyroidism is now rare, but we are aware of two patients who died of severe undiagnosed and untreated thyrotoxicosis in Chicago within the past few years (259). Deaths most frequently are attributed to cardiovascular complications such as myocardial infarction, arrhythmia, or heart failure, or infections secondary to debility. Some patients become spontaneously hypothyroid, and in fact most individuals apparently cured of Graves' thyrotoxicosis demonstrate evidence of hypothyroidism decades later. Coincident autoimmune thyroiditis presumably plays a role in such thyroid atrophy. Since in some patients treatment of thyrotoxicosis is associated with the spontaneous reestablishment of thyroid homeostasis after a period of enforced reduction in hormone formation (by drugs, surgery, or 131I treatment), it is obvious that the thyrotoxic phase of the disease can be self-limiting.

Toxic crisis, or thyroid storm, was also a frequent feature of Graves' disease in the past. This serious and often fatal development was a marked accentuation of the thyrotoxicosis, with hyperthermia, uncontrolled tachycardia, weakness, and delirium. This situation, now rarely encountered, is discussed in Chapter 12.

The ophthalmopathy of Graves' disease may follow a course quite different from that of thyrotoxicosis. This topic is also discussed in Chapter 12.

SYMPTOMS AND SIGNS OF GRAVES’ DISEASE AND THYROTOXICOSIS

In patients with Graves' disease, the ocular changes, lymphoid hyperplasia, localized abnormalities of skin and connective tissue (e.g., acropachy) and the goiter itself represent parts of the autoimmune syndrome. The remainder of the changes appear to be entirely attributable to an excess of thyroid hormone. Certain systems or organs (e.g., the muscles and cardiovascular system) play paramount roles in the disease, but as far as can be determined, these changes are all fundamentally related to and dependent on the excessive serum concentration of thyroid hormones.

Often the presenting symptoms are weight loss, weakness, dyspnea, palpitations, increased thirst or appetite, hyperdefecation, irritability, profuse sweating, sensitivity to heat or increased tolerance to cold, or tremor. Occasionally, prominence of the eyes or diplopia is the apparent symptom, and goiter may long antedate all other manifestations. Often a relative or friend notices eye signs, goiter, or nervous phenomena before the patient is conscious of any departure from his or her usual status. This asymptomatic phase of thyrotoxicosis is more commonly found in men and children. The excess of thyroid hormone produces an intoxication that in some persons takes the form of exhilaration. They may feel not only healthy but healthier than usual at a time when they are displaying unmistakable objective evidence of thyrotoxicosis. In older patients particularly, the symptom or symptoms may point to the heart more than to any other part of the body, "thyrotoxicosis masquerading as heart disease."

The habitus in Graves' disease shows nothing characteristic. In childhood, those afflicted are tall for their age. This association is an effect of the disease, not an etiologically related variable.

The nutritional state varies greatly. Sometimes the patient is severely emaciated, but on average the weight loss is 5 - 20 lbs. Infrequently, perhaps in 1 out of 10 instances, the patient actually gains weight while thyrotoxic.

The face may may instantly provide the diagnosis. An expression of fright or extreme anxiousness is common, largely because of the peculiar eye signs that may be present. Marked flushing is often noted. A drawn or sunken appearance may result from emaciation or dehydration. It is possible, especially in older patients, to find a considerable degree of thyrotoxicosis without any distinguishing evidence in the facies.

A change in reaction to external temperature is a very classic symptom. The development of a preference for cold weather, of a desire for less clothing and less bed covering, and of decreased ability to tolerate hot weather is highly suggestive of hyperthyroidism.

The tongue tends to be red and smooth; it may also exhibit a definite tremor. The tonsils, if present, are usually rather large, as is the postpharyngeal lymphoid tissue.

The neck is usually conspicuous due to the goiter. It is possible, although rare, for thyrotoxicosis to exist without a visible or palpable goiter. We note reports in the literature that up to a quarter of patients may not have a goiter [272], but this is not our experience. In the neck, the carotids will often be seen to throb violently; this condition may contribute to the anxiety of the patient.

The eye signs characteristic of Graves' disease often constitute the most striking feature (Fig. 10-4 above, Fig. 10-5, Fig. 10-6 below). Prominence of the eyes is the most important sign. A wild or staring expression is often observed. Lag of the lids behind the globes on downward rotation and lag of the globes behind the lids in upward rotation, infrequent blinking, failure to wrinkle the forehead on looking upward, and decreased ability to converge are also cardinal manifestations. Swelling of the lids is a characteristic and frequent eye sign. The bulbar conjunctiva may be edematous (chemosis). The insertions of the medial and lateral rectus muscles are often enlarged, inflamed, and quite obvious . The lacrimal gland can become infiltrated by lymphocytes and enlarged, and may protrude below the orbital boney margin.

Figure 5a. This MRI image from a patient with Graves' ophthalmopathy provides a coronal view of the eyes. In this depiction the muscles appear white, and are enormously enlarged, especially in the left eye.

Figure 5b. In this transverse view the enlarged muscles are seen (appearing dark against the light fat signal) and the exophthalmos is apparent.

Figure 6a. Histologic appearance of extraocular muscle.

Figure 6b. Histologic appearance of retrobulbar fat.

Figure 6c. Histologic appearance of lacrimal gland removed during a Kronlein procedure on a patient with severe exophthalmos.

For convenience, the ophthalmic phenomena may be grouped as in Table 10-4. A classification of the eye changes and a system of grading of their severity have been adopted by the American Thyroid Association [273] and is given in Chapter 12. 

Table 4. Ocular Signs and Symptoms in Graves' Disease

 Ophthalmic phenomena reflecting thyrotoxicosis per se and apparently resulting from sympathetic overactivity:

Lid reaction

•  Wide palpebral aperture (Dalrymple's sign)

•  Lid lag (von Graefe's sign)

•  Staring or frightened expression

•  Infrequent blinking (Stellwag's sign)

•  Absence of forehead wrinkling on upward gaze (Joffroy's sign)

 

Ophthalmic phenomena unique for Graves' disease and caused by specific pathologic changes in the orbit and its contents:

•  Inability to keep the eyeballs converged (Mobius' sign)

•  Limitation of movement of the eyeballs, especially upward

•  Diplopia

•  Blurred vision due to inadequate convergence and accommodation

•  Swelling of orbital contents and puffiness of the lids

•  Chemosis, corneal injection, or ulceration

•  Irritation of the eye or pain in the globe

•  Exophthalmos (also produces mechanically a wide palpebral fissure)

•  Visible and palpable enlargement of the lacrimal glands

•  Visible swelling of lateral rectus muscles as they insert into the globe, and injection of the overlying vessels

•  Decreased visual acuity due to papilledema, retinal edema, retinal hemorrhages, or optic nerve damage

The eye signs may vary independently of the intensity of the thyrotoxicosis. Although it is true that in most patients with Graves' disease, eye signs, goiter, and symptoms of thyrotoxicosis appear more or less coincidentally, it is also true that in certain cases eye signs may appear long before thyrotoxicosis is evident, or become worse when the thyrotoxicosis is subsiding. Indeed, in some patients, serious exophthalmos may develop at a time when the thyrotoxicosis has been controlled by treatment.

The eye symptoms are extremely distressing. Diplopia is common; decreased visual acuity and other visual disturbances are less common. More frequent are symptoms due to conjunctival or corneal irritation. These symptoms include burning, photophobia, tearing, pain, and a gritty or sandy sensation.

Horner's syndrome on one side is occasionally encountered when the goiter has pressed upon the trunk of the cervical sympathetic chain. This syndrome consists of unilateral enophthalmos, ptosis of the lid, and miosis, as well as decreased sweating on the homolateral face.

THYROID GLAND

The thyroid may be smooth, lobulated, or rarely nodular. In thyrotoxicosis associated with nodular goiters, the hyperfunctioning tissue may reside between the nodules [274], which would constitute Graves' disease in a nodular goiter. Often the surface is lobulated, and the upper poles may seem to contain nodules above the site of entry of the superior thyroid artery. The diffuse toxic goiter is usually more or less symmetric. The size is related, but not closely, to the severity of the disease. It varies from the barely palpable normal (15 - 20 g) to an enlargement of six times normal (100 g) or, rarely, even more, but averages about 45 g. The near symmetry and usually moderate size of the diffuse goiter of Graves' disease make it somewhat less unsightly than many of the nodular goiters. It is commonly stated that the gland is not palpable in 1% of cases, either because the thyroid is actually smaller than ususal or because it is beneath the manubrium. However, in the presence of thyrotoxicosis, a small or normal-sized thyroid should alert the physician to the possiblity of some other cause of the illness.

The consistency of diffuse toxic glands is firm but elastic, or very firm if iodide has been given. The borders are easily demarcated by palpation. The pyramidal lobe should always be searched for since enlargement indicates the presence of diffuse disease of the thyroid. Also, if left behind at operation, it may be the site of recurrence of the disease.

Thrills and bruits are important but often absent. Their presence usually denotes hyperfunction. A thrill is less common than a bruit. It is more likely to be felt as a systolic purr in the region of the superior poles over the superior thyroid arteries. Bruits may be continuous or systolic in time, similar to a blowing cardiac murmur. Usually they are audible over the entire thyroid, often being louder on one side than on the other. Either a thrill or a bruit is highly suggestive, but not pathognomonic, of thyrotoxicosis. If local examination of a goiter discloses either of these signs, even though other evidence of hyperfunction may be lacking, especially careful investigation into the possibility of thyrotoxicosis is indicated. Both thrills and bruits tend to decrease in intensity as thyrotoxicosis subsides. They completely disappear in a few days under treatment with iodide.

The thrill is the palpable and the bruit the audible sign of turbulence associated with an increased rate of flow through rather tortuous vessels. The location of the thrill suggests that the larger thyroid vessels are chiefly responsible. Bruits may be distinguished from venous hums by occlusion of venous return caused by gentle pressure above the thyroid. A carotid or innominate thrill or bruit may be difficult to distinguish from sounds originating in the thyroid gland. Their localization over the vessel and distal transmission usually allow a distinction to be made.

Neighborhood symptoms, including dysphagia and the sensation of a lump in the neck, may be produced by toxic as well as nontoxic varieties of goiter. Sometimes the supraclavicular lymph nodes become enlarged and tender [275].

Vocal cord palsy is encountered, but is found chiefly in cancer of the thyroid, occasionally in nodular goiter, and only rarely in Graves' disease. Occasionally it is found on routine preoperative laryngoscopic examination, having produced no symptoms such as dysphonia or hoarseness.

The Skin

Cutaneous manifestations are nearly always present when hypermetabolism is significant. The patient feels hot and prefers a cold environment. Active sweating occurs under circumstances that would provoke no response in normal persons. Hand shaking gives a nearly diagnostic impression. The hand of the thyrotoxic person is erythematous, hot, and moist (sometimes actually dripping wet), in a state of hot hyperhydrosis. Although such hands may occasionally be found in other conditions, the finding of a cold hand, dry or moist -- almost excludes hyperfunction of the thyroid. Flushing is also very common, more in younger patients than in older ones. There may be more or less continuous erythema of the face and neck, with superimposed transient blushing. Occasionally diffuse pruritis or urticaria occurs. Urticaria appears to be linked to Graves hyperthyriodisim by some immune mechanism, but so far the causal relation is unknown (276).

The vasomotor system is overactive. Many of these cutaneous manifestations may be considered expressions of or incidental to increased heat elimination.

Redness of the elbows, first noted by Plummer, is frequently present. It is probably the result of the combination of increased activity, an exposed part, and a hyperirritable vasomotor system.

Although the integument is thinned, manifestations due to alteration in the growth of the tissue are less evident. It is possible that the type of fingernail described by Plummer (onycholysis) belongs in this category (Figure 10-7). The process may involve all fingers and toes, but typically begins on the fourth digit of each hand. The free margin of the nail leaves the nail bed, producing a concave or wavy margin at the line of contact. The hyponychium may be ragged and dirty, despite the best efforts at personal hygiene. Plummer's nails are a frequent and interesting clinical finding in Graves' disease. Occasionally the spoon-shaped fingernails of hypochromic anemia are encountered.

Figure 7. Plummer's nail changes, showing thinning of the nail and marked posterior erosion of the hyponychium.

Patchy hyperpigmentation, especially of the face and neck, is frequently seen, and occasionally there is a general increase in pigmentation. Most dark-skinned persons detect a definite increase in pigmentation during the onset of thyrotoxicosis, which may be dramatically localized around the eyes. 

Patchy vitiligo is found in 7% of patients with Graves' disease, and we have observed several instances of complete loss of pigmentation in association with thyrotoxicosis. These changes are manifestations of associated autoimmunity directed toward melanocytes. The vitiligo, often of the hands and feet, may precede the onset of Graves' disease by years or even decades. Observation of this change is a useful clinical sign when attempting to establish the cause of thyrotoxicosis or exophthalmos.

Hair tends to be fine, soft, and straight. Women may complain that it will not retain a curl. (This complaint is also typical of patients with myxedema.) Temporary thinning of the hair is common, but alopecia is rare. Hair loss is often extreme after marked changes in metabolic rate are induced during therapy. We have seen complete or partial alopecia develop in a few patients with Graves' disease, sometimes in association with urticaria. These changes are believed to be manifestations of autoimmunity directed against the hair follicles.

Peripheral edema, unrelated to congestive heart failure or renal disease, is very common.

Pretibial myxedema (Figure 10-8) and the other remarkable abnormalities of "thyroid acropachy" are discussed in Chapter 12.

Figure 8. Remarkable "pretibial myxedema", also present on feet and hands, of a patient with Graves' disease and exophthalmos.

NEURAL AND MENTAL CHANGES

Neural and mental findings are varied and striking. The patient complains of nervousness or irritability and appears to be restless and fidgety. It sometimes seems impossible for the thyrotoxic patient to remain still for an instant. The tendon reflexes tend to be brisk, and the reflex relaxation time is shortened. The reaction to all sorts of stimuli is distinctly excessive. When asked to sit up, the patient jumps into an upright position. He or she may wish to cooperate but rather overdoes it. The patient is, so to speak, "hypercooperative." Such behavior constitutes an almost pathognomonic pattern. In the clinic, we are familiar with what we call the "thyrotoxic entrance and exit." The thyrotoxic patient hops into the clinic room like a jack-in-the-box, often with staring eyes, sits very quickly in the clinic chair, bolt upright, looks rapidly about the room, and does whatever is asked with pathologic alacrity. His or her exit is equally precipitate. Often emotional instability is combined with this pattern, perhaps to the point of a significant change in personality. The patient is often given to fits of crying, but may have sufficient insight to realize that the crying is pathologic. Some patients become hyperirritable and combative, and this can precipitate accidents or even assaultive behavior.

In some patients, the emotional pattern is that of hypomania or pathologic well-being (euphoria). In others, hyperactivity seems to produce a state of exhaustion, and profound fatigue or asthenia chiefly characterizes the picture. The mind is often very active, and the patient is troubled with insomnia. Rarely, patients develop visual or auditory hallucinations or a frank psychosis. The latter may not completely clear up after thyrotoxicosis has been treated. It is probable that thyrotoxicosis makes manifest an abnormality already present rather than inducing a psychosis de novo. Brownlie et al reported 18 cases of patients with thyrotoxicosis who had coincident psychotic disorder and concluded that, usually this was an affective psychosis, and that the incidence was above chance co-occurrence. Thyrotoxicosis appeared to be a precipitant effect of psychosis [277].

Impairment of intellectual function has been found in patients with untreated hyperthyroidism. It is usually assumed that such abnormalities return to normal with therapy. However, Perild et al. [166]report that ten years after successful therapy of thyrotoxicosis a group of patients manifested abnormal neuropsychological tests, and half had significant intellectual impairment which was apparently permanent. This surprising observation awaits confirmation. Marked increase in fatigability, or asthenia, is often prominent. This increased weariness may be combined with hyperactivity. Patients remark that they are impelled to incessant activity, which, however, causes great fatigue.

A fine, rapid tremor of the outstretched fingers is classically found, and a generalized tremulousness, involving also the tongue, may be evident. Muscle fibrillations are not a usual part of the syndrome, but they may occur in chronic thyrotoxic myopathy. Polyneuropathy has also been reported. [279]

More severe neurologic problems also occur during Graves' disease ( Table 10-5). Patients who are known to have a convulsive disorder may become more difficult to control with the usual medications, and seizures may appear in patients who have never previously manifested such symptoms [280]. Electroencephalography [280] reveals increased fast wave activity, and occasionally bursts of tall spike waves. Several reports describe a severe steroid responsive encephalopathy (Hasshimoto’s Encephalopathy) in patients with Hashimoto's thyroiditis ( 281). The same syndrome has been described in Graves' Disease [282]. A  direct relation to Graves, or thyroiditis, seems probable, but is un-proven. In animals, excess T4 decreases the threshold to convulsive stimuli [169].

 

Table 5. Neuromuscular Manifestations of Thyrotoxicosis

Tremor

•  Hyperactive reflexes

•  Accelerated reflex relaxation

•  Anxiety

•  Disorientation

•  Psychosis

•  Thyrotoxic neuropathy (rare)

•  Acute thyrotoxic encephalopathy ( rare)

•  Seizures (with or without an underlying abnormality)

•  Neuropathy secondary to nerve entrapment by lesions of pretibial myxedema

•  Corticospinal tract disease with pyramidal tract damage (rare)

•  Chorea and athetoid movements (rare)

•  Hypokalemic periodic paralysis

•  Myopathy

•  (Myasthenia gravis -- associated)

 

 

C.H., 52-Year Old Woman: Psychosis with Thyrotoxicosis

This woman appeared in the emergency room in a confused and agitated state. She refused to talk, but would on occasion answer questions. She appeared to be extremely paranoid and was resistant to offers of help.

She came to the emergency room alone, and after one interview disappeared. She returned a few hours later, again in the same agitated, confused, paranoid, and semimute condition. She stated that she heard voices quoting the Scripture and denied that these voices directed her to harm herself or others, but indicated that she was responsible for the bad problems of the world.

Relatives were contacted and indicated that the patient had been entirely well up to the previous few days, when she had become confused and agitated. It was determined that the patient had worked for more than 20 years and had lost her position about four years previously. She was married and had been separated from her husband intermittently during the past four years. She knew her address, was aware of the month and year but not the date, and was confused about current events.

The BP was 150/80 and the pulse rate 140. The patient was disheveled, thin, and hyperactive. The eyes were normal. Results of routine blood chemistry tests, complete blood count, and urinalysis were negative.

The patient was treated initially with haloperidol (Haldol), 1 mg twice a day, and gradually calmed. The diagnosis of hyperthyroidism was considered and confirmed by an FTI of 19. Antithyroid antibodies were absent.

During treatment, the patient's paranoia and anxiety subsided. She subsequently indicated that there had been a gradual increase in tiredness and weakness, weight loss of 10 lbs, heat intolerance, palpitations, and tremor over one to two years. Previous medical problems included a hysterectomy for fibroids and mild hypertension treated by diuretics. There was no history of previous psychiatric illness in the patient or her family.

On further examination, the thyroid was seen to be enlarged to about 35 g and was diffusely increased in size, without nodules; there was no bruit. Propranolol was added to the therapy, and Haldol was continued. The patient rapidly became psychologically normal and entirely cooperative, and regained control of her personal affairs.

An RAIU test was 49.7%. The patient received 4 mCi of radioactive 131I. After radioactive iodine therapy, the patient was given an antithyroid drug that brought her thyroid hormone levels back to normal. When this drug was discontinued, her FTI returned to 15.7. She was given 3.4 mCi of radioactive iodine again, and PTU was restarted. When last examined, her FTI was in the normal range.

There has been no return of any abnormal psychologic function, and the patient has received no further psychiatric care.

This episode appeared to be an acute psychotic reaction associated with severe hyperthyroidism, occurring in a patient with no previously known psychologic disease. It cleared promptly with medical therapy, including treatment of the hyperthyroidism, and the patient is now apparently well.

C.J., 43-Year-Old Woman: Thyrotoxic Neuropathy

This woman was referred for evaluation with a history of obesity, hypertension for two years, prominence of the right eye for two years, and thyroid overactivity known for six months. She had gained 50 lbs during the interval preceding the examination because of excess eating. Increasing dyspnea and shortness of breath, present for the previous two years, had become worse in the previous two months. She came to the emergency room because of symptoms of asthma. Examination revealed a pulse rate of 120 and an enlarged heart. There was LVH and strain on the electrocardiogram, and on echocardiogram an enlarged left atrium and a left ventricle with decreased function, especially of the lateral and posterior inferior walls. Thyroid function tests showed a T4 level of 17 ug/dl, an FTI of 16.8, and a T3 level of 357 ng/dl. She received digoxin, 0.25 mg daily, furosemide, 40 mg daily, potassium chloride, and aminophylline.

On examination in the endocrine clinic, the BP was 170/100, and the patient was obese and hyperactive. There was moderate bilateral proptosis and inflamed insertions of the extraocular muscles. There was 22 mm proptosis bilaterally. The thyroid was diffusely enlarged to about 40 g. Neurologic examination showed weakness of ocular motility with diplopia on the left lateral gaze, bilateral nystagmus, marked proximal muscle weakness without fasciculations, and decreased touch, pinprick, and vibration sense in a glove distribution of both arms, the left greater than the right. There was no significant deficit in the feet. Weakness in the left upper extremity was marked. Deep tendon reflexes were absent. The differential diagnosis included Graves' disease, cardiomyopathy and peripheral neuropathy, congestive heart failure, and hypertension.

A neurologic consultant confirmed the neuropathy and noted mild choretic movements of the left hand and arm. Other known causes of neuropathy were excluded. The patient was treated for one month with antithyroid drugs and then given 2.7 mCi 131I. Because of continued hyperthyroidism, the patient was retreated with 3.2 mCi 131I seven months after the initial treatment. Three months later the FTI was normal at 10.4, and there were no symptoms or signs of congestive heart failure. Some decreased strength and clumsiness of the left hand persisted. The diplopia and proptosis were unchanged. The neuropathy in the hands had decreased, and the patient was euthyroid.

This patient exhibited profound cardiomyopathy and skeletal myopathy, choreiform movements, and peripheral neuropathy, all apparently related to severe thyrotoxicosis. She improved rapidly with appropriate treatment of the thyrotoxicosis.

The tremor of Parkinsonism is greatly intensified during thyrotoxicosis. Signs and symptoms of cerebellar disease or pyramidal tract lesions have been seen [170,171]. Rarely, patients manifest extreme restlessness, disorientation, aphasia, grimacing, chorioathetoid movements, symptoms suggestive of encephalitis [286], or episodes of hemiparesis or bulbar paralysis. These symptoms clear up completely after restoration of metabolism to normal. No definite lesions have been found in the brain. Rarely, polyneuropathy has been severe enough to cause paraplegia [287].

Most of the biochemical actions of thyroid hormone on the brain are related to developmental functions rather than function in the adult. These actions have recently been reviewed by Bernal.  All three forms of thyroid hormone receptor are expressed in the brain, especially in neurons. Genes regulated by thyroid hormone include myelin basic protein, mitochondrial genes such as cytochrome C oxidase, neurotrophins and their receptors, including NGF and trkA, cytoskeletal components such as tubulin, transcription factors such as NGF1a, extracellular matrix proteins, and adhesion molecules such as NCAM, genes involved in intracellular signaling such as RC3/neurogranin, and genes expressed in the cerebellum such as pcp-2. Interestingly, the brain of a thyrotoxic human subject does not have an elevated consumption of oxygen. Sensenbach et al. [174] found the cerebral blood flow to be increased, the cerebral vascular resistance decreased, arteriovenous (AV) oxygen difference decreased, and oxygen consumption unchanged in thyrotoxicosis. Reciprocal changes occurred in myxedema, and all reverted to normal after therapy. Curiously, brain size was shown to decrease significantly during treatment of the hyperthyroid patients, and ventricular size increased. This remarkable change is of uncertain cause but may involve osmotic regulation.

Although it is possible that some of the central nervous system irritability is a manifestation of elevated sensitivity to circulating epinephrine, this contention has not been proved. Epinephrine levels and catecholamine excretion are actually not elevated, but propranolol, presumably acting by inhibition of alpha-adrenergic sympathetic activity, certainly reduces anxiety and tremulousness in a very useful manner. The clinical applications of these findings are discussed in Chapters 11and 12.

MUSCLES

The muscular symptoms vary from mild myasthenia to profound muscular weakness and atrophy, especially of proximal muscle groups. This weakness forms the basis of a useful clinical test. If a thyrotoxic patient seated in a chair is asked to hold one leg out straight and in a horizontal position, he or she may be able to do so for 25 - 30 seconds only; normal persons can maintain such a position for 60 - 120 seconds. Toe standing and step climbing may also bring out muscle weakness that is otherwise not so apparent. In the more extreme forms of muscular involvement, there is not only weakness but also atrophy. Wasting of the temporals and interossei may be noted in a considerable number of patients, and in a few, wasting of all skeletal muscles. This wasting may go so far as to bear a close resemblance to progressive muscular atrophy; occasionally the myopathy may shade into the picture of a polymyositis. Muscle cell necrosis and lymphocyte infiltration may be visible histologically, but usually are not found even when the symptoms of weakness are severe [175]. Tremor, which is usually present, is ascribed to altered neural function. Fasciculations are unusual.

The speed of both tension development [290] and relaxation of the muscles is increased, so that the reflex time is shortened. The electromyogram is normal in most instances but may occasionally resemble that of muscular dystrophy [291]. Work efficiency, measured in terms of the calories of heat produced while performing a given amount of work, has been reported to be both decreased [292] and normal. The question of metabolic efficiency of hyperthyroid muscle has been revisited by Erkintalo et al, using phosphorus-31 MRI spectroscopy, finding that toxic muscle required more energy to function than normal, presumably because of additional ATP-consuming mechanisms [293]. Creatine excretion is increased. The muscles have decreased ability to take up creatine, produced in the liver, from the blood [294-295]. Creatinine excretion is initially increased by the general catabolism of hyperthyroidism, but as muscle mass diminishes, creatinine excretion in the urine is depressed.

Myasthenia gravis may simulate thyrotoxicosis, and vice versa [296]. It has been reported that neostigmine both strengthens the muscles in thyrotoxic myopathies and is without effect. Certainly, the response is small in comparison with the immediate and striking correction of weakness seen in myasthenia gravis. Thyrotoxicosis may rarely ameliorate myasthenia gravis, but typically it is accentuated by thyrotoxicosis and is also worsened by myxedema. The close relationship between these two diseases is apparent in the observation that thyrotoxicosis occurs in 3% of patients with myasthenia gravis. The pathogenic anti-acetylcholine receptor antibodies that occur in myasthenia gravis are clearly comparable to the anti-TSH receptor antibodies found in Graves' disease. In addition, it has been found that TG and acetyl- cholinesterase share epitopes recognized by B cells. It is, however, uncertain that this plays any role in the pathogenesis of muscle disease in Graves' patients.

Periodic paralysis is precipitated and worsened by thyrotoxicosis [297]. This relationship has been extensively studied in Japan, where it is a familiar syndrome, particularly in men. The paralysis is usually associated with and due to hypokalemia. While the exact mechanism is not known, the hypokalemia is believed to be caused by a shift to the intracellular compartment. It has been demonstrated that thyrotoxicosis augments K+ uptake and release from cells. Experimental T4 treatment augments synthesis of membrane Na+-K+ activated ATPase. The episodes of paralysis tend to be infrequent and sporadic, but most commonly occur after a meal, following exercise, or start during sleep, and can be induced by administration of glucose and insulin. The onset following meals or exercise presumably relates to rapid K+ uptake by cells. Episodes last from minutes to hours, usually involving peripheral muscles, but can cause paralysis of the diaphragm and affect the heart. Serious episodes can be associated with extensive muscle cell damage and necrosis, EKG abnormalities such as ST and T wave changes, PVCs, first degree heart block, prolonged Q, T intervals, and even ventricular fibrillation [298].

Potassium treatment has some protective effect, and quickens recovery from attacks. Propranolol, for reasons not entirely clear, has prophylactic action. Therapy of the thyrotoxicosis almost always causes the rapid and permanent disappearance of the syndrome.

Myotonia congenita and myotonia dystrophic do not occur with increased frequency with thyrotoxicosis.

SKELETON AND CALCIUM METABOLISM

Roentgenographic examination of the bones frequently discloses evidence of decalcification. Microdensitometry demonstrates this condition at all ages and in both sexes [299-301]. Patients with even mild increases in thyroid hormone lose bone mass [302], especially if postmenopausal and not receiving estrogen therapy. Those with a history of thyrotoxicosis extending over a number of years may have osteoporosis that is severe and premature9303-304. Fractures are uncommon, with the most frequent being collapsed vertebra in a chronically thyrotoxic postmenopausal woman. Skeletal mass is augmented after therapy [300-301]. Treatment restores the density in younger patients, but not usually in the elderly [300]. Although most attention has been made to the effects of thyroid hormone on bone density in women, it is not surprising to know that thyroid hormone excess also has a mild deleterious effect in males (303). A meta analysis of 289 published studies on the effect of hyperthyroidism causing bone fragility found that hyperthyroid patients had decreased bone mineral density and increased fracture risk. The bone mineral density tended to return to normal after therapy (302).

Periarthritis of the shoulder (subacromial bursitis) is occasionally associated with thyrotoxicosis. Linear bone growth may be accelerated in children. The time of epiphyseal closure may be accelerated in children, and bone age may exceed chronologic age.

Thyrotoxicosis results in an accelerated turnover of bone calcium and collagen [305-306]. TRα1, TRβ1, and TRβ2 proteins are expressed in human osteoblast cells and strongly in human bone marrow stromal cells. Endogenous receptors in these cells are functional in in vitro test systems. The specific function in vivo is unclear. As described in the section on pathology, the histologic picture of bones from the thyrotoxic patient may suggest osteitis fibrosa with increased osteoclastic activity, fibrosis, and an increased number of osteoblasts [267]. Histomorphometric evaluations with tetracycline labelling demonstrates accelerated bone resorption and formation, both in spontaneous hyperthyroidism and in women treated with excess thyroid hormone [266, 306,305].  In bone biopsy specimens the thin trabeculae of osteoporosis are seen. [267]The serum calcium level is usually normal, but may be elevated sufficiently to produce nausea and vomiting [307]and, rarely, renal damage [308, 309]. It may be made clinically evident when thyrotoxic patients become relatively immobile, for example at bed rest during illness. In contrast to what occurs in hyperparathyroidism, the hypercalcemia can usually be corrected partially or totally by the administration of glucocorticoids [310], but these have not been effective in all cases [311]. Phosphorus administration also lowers the concentration of calcium in serum and urine to normal [312]. The exchangeable calcium pool is remarkably increased [305]. Serum osteocalcin is increased in parallel with hormone levels [313]. The alkaline phosphatase level may be elevated, with a pattern showing the normal equal distribution of bone and liver isoenzymes. The changes in calcium and alkaline phosphatase correlate with serum T3 levels [314]. After therapy, the alkaline phosphatase level tends to increase, and bone isoenzyme becomes predominant, probably due to skeletal repair [314].

Fecal and urinary calcium excretion is greatly augmented, and it is remarkable that renal stones are rarely formed. This is because there is a concomitant increase in excretion of colloids that stabilize the calcium. Urinary hydroxyproline and pyridium cross-link excretion are increased and fall to normal after therapy [306]. Serum carboxy-terminal-1-telopeptide and serum osteocalcin levels and urinary osteocalcin secretion are increased and return to normal with therapy[268,313,315].

The serum phosphorus level is in the normal range or depressed. Renal phosphorus resorption is in the normal range or elevated [309, 310]. Although some of the observations suggest the presence of hyperparathyroidism, it is most likely that the changes actually reflect the direct metabolic effects of thyroid hormone. The parathyroid glands are histologically normal. In fact, parathyroid hormone (PTH) levels tend to be suppressed in hyperthyroidism, apparently in response to the elevated calcium levels [201]; 1,25-dihydroxyvitamin D3 levels are likewise about 40% below normal [202].

The increased fractional tubular phosphate reabsorption characteristic of hypoparathyroidism may also occur in thyrotoxicosis, probably because of reduced PTH levels. In one reported study [308], urinary phosphorus excretion was depressed after calcium infusion. Thus, a normal response was obtained rather than that found in hyperparathyroidism.

The hypercalcemia appears to be a direct manifestation of thyroid hormone action on bone metabolism [316,317], and calcium absorption from the intestine is usually reduced. [318]Both catabolism and anabolism of bone are accelerated. Negative calcium balance can sometimes be corrected by administration of calcium, an observation that perhaps should be given more attention in the management of thyrotoxic patients. Hypercalcemia can be corrected by propranolol therapy in some patients [319]. Bone turnover can be reduced by pamidronate and by calcitonin, which may therefore have a useful role in reducing thyrotoxicosis-induced osteopenia [320-321].

Two exceptional cases have been reported with coincident thyrotoxicosis and hypercalcemia with elevated PTH levels. Treatment of thyrotoxicosis eliminated all abnormalities, for reasons unknown. [322].

RESPIRATORY SYSTEM

Except for dyspnea, which may or may not represent abnormal respiratory function, symptoms deriving from the lungs are not prominent. Nevertheless, measurements show some reduction in vital capacity, expiratory reserve volume, pulmonary compliance, airway resistance, and weakness in both expiratory and inspiratory muscles [323,324]. Minute volume response to exercise is excessive for the amount of oxygen consumed [325]. Dyspnea on effort is present in a large majority of the patients. Pulmonary function in patients without coincident congestive heart failure has demonstrated reduction in vital capacity, decreased pulmonary compliance, weakness of the respiratory muscles, increase in respiratory dead space ventilation, and normal diffusion capacity. A combination of the four abnormalities may produce dyspnea [326]. Amelioration of intractable bronchial asthma has been reported after treatment of coincident thyrotoxicosis [209].

CIRCULATORY SYSTEM

(Table 10-6)

First and foremost of the symptoms deriving from the circulatory system are palpitations and tachycardia. The heart may beat with extreme violence, which may be distressing to the patient, particularly at night or on exercise. The pulse on palpation is rapid and bounding. The systolic blood pressure is frequently elevated. The diastolic blood pressure is characteristically decreased, and the pulse pressure is elevated, being usually between 50 and 80 mm Hg.

Left ventricular hypertrophy may be suggested on physical examination. A bounding precordium is so typically found that its absence is a point against the diagnosis of hyperthyroidism. However, in the majority of instances, roentgenograms show the heart to be normal in transverse diameter. A systolic murmur is usually heard over the precordium. One reason for this murmur is the development of mitral valve prolapse during thyrotoxicosis. [328]This can be detected by angiography, or more easily by echography. It is postulated that papillary muscle dysfunction due to inadequate ATP supplies may be responsible for the lesion. Prolapse is usually not clinically evident but rarely is a cause of symptomatic mitral valve insufficiency. The prolapse can revert to normal with therapy [328].

Interpretation of physical signs, especially systolic murmur and gallop rhythm, in the heart is difficult and uncertain in the presence of thyrotoxicosis. In evaluating heart status in thyrotoxicosis, one should concentrate chiefly on the presence or absence of signs of failure rather than on physical signs in the heart itself. If no signs of failure are present, the best procedure regarding abnormal cardiac findings is to ascertain whether they persist after thyrotoxicosis has been abolished.

A grating pulmonic systolic sound ("Lerman Scratch"), which has some of the characteristics of a pericardial friction rub, is occasionally heard over the sternum in the second left interspace. It is heard best at the end of full expiration. Its intensity subsides as thyrotoxicosis improves. The diagnosis of pericarditis may be suggested on the basis of this sound. The fact that it is superficial and tends to disappear on inspiration suggests a pleuropericardial origin. It may be related to the dilated pulmonary conus often seen on x-ray films in thyrotoxic patients. The sign has no prognostic significance.

Extrasystoles are frequent, and paroxysmal atrial tachycardia and atrial fibrillation, paroxysmal or continuous, occur in 6 - 12% of patients. Even subclinical hyperthyroidism is associated with a fivefold greater chance of developing atrial fibrillation, and it is effectively the same as the situation with overt hyperthyroidism (329). Precordial pain that seems distinct from angina pectoris occurs occasionally. Cardiac enlargement and congestive heart failure may occur with or without prior heart disease [331, 332]. (Figure 10-9). The electrocardiographic manifestations are confined to tachycardia, increased voltage, and sometimes a prolongation of the PR interval [332], unless there is a dysrhythmia or an accompanying but unrelated disorder of the heart.

Figure 9. Congestive heart failure induced in an otherwise healthy young woman (a), which receded (b), and returned to normal (c), during and after therapy.

Patients with coronary atherosclerosis often develop angina during thyrotoxicosis. Occasionally angina develops de novo in young women with arteriography-proven normal coronary arteries. This condition has been ascribed to an imbalance between increased cardiac work and blood supply, so that a functionally deficient blood supply occurs even with a patent vessel [333]. Severe coronary vasospasm has been observed during angiography in patients with GD (334). Myocardial damage occurs in toxic patients with CHF (335) and myocardial infarction can occur in toxic patients with normal coronary vessels [336]. 

In thyrotoxicosis the heart rate, stroke volume, and cardiac output are increased. Circulation time is decreased. There is dilatation of superficial capillaries. Coronary blood flow and myocardial oxygen consumption in each stroke are increased [337]. Circulating plasma volume is increased. AV oxygen differences are variable but tend to be normal. Cardiac output in response to exercise is excessive in relation to oxygen consumed.

The relation of cardiac systolic time intervals to thyroid function has provided a valuable in vivo bioassay of hormone action. The pre-ejection period is shortened in thyrotoxicosis, and the left ventricular ejection time remains relatively normal. The interval from initiation of the QRS complex to arrival of the arterial pulse in the brachial artery is reduced [338]. Cardiac diastolic function as evaluated by echocardiography remains normal in the majority of patients [339].

Congestive heart failure and atrial fibrillation, when due to or associated with thyrotoxicosis, are relatively resistant to the action of digoxin. Accelerated metabolism of digoxin, plus the cardiac inefficiency and irritability produced by thyrotoxicosis, may be at least two of the factors producing this resistance. Although the response to the drug will be blunted, a beneficial effect will occur if a proper level of digoxin is attained.

Atrial fibrillation should be treated by anticoagulation if it is persistent, since it is associated with serious embolism in 10% of cases. The usual contraindications of old age, HBP, bleeding tendency, recent CVA, etc. apply, and the dose of Coumadin needed is lower than normal in thyrotoxic patients. AF tends to revert spontaneously to normal when hyperthyroidism is cured, but this may not occur before six months, or not at all, if AF was of long standing. Therapeutic cardio- version is recommended if AF persists six months beyond achievement of euthyroidism. Long term follow-up studies have revealed increased mortality from cardiovascular and cerebral vascular disease in patients with a past history of overt hyperthyroidism treated with radioiodine, and in patients with subclinical hyperthyroidism. Possibly development of atrial fibrillation and other supraventricular dysrhythmias may account for increased vascular mortality (340). Treatment of thyrotoxic heart disease has been reviewed recently [341]. A review of the impact of hyperthyroidism on cardiac function in older patients is available [342].

It has been suggested that the changes in the cardiovascular system are secondary to increased demand for metabolites and to increased heat production. Dilatation of superficial capillaries for the dissipation of heat does cause increased blood flow and cardiac output. However, a direct action of thyroid hormone on the heart is also increased, since the sinus node has higher intrinsic activity, the isolated thyrotoxic heart beats faster than normal, and isolated papillary muscle from a thyrotoxic heart has a shortened contraction time [343-345]. The heart shares in the general increase in respiratory quotient found in skeletal muscle. Adenosine transport into myocardial cells and its phosphorylation are increased [345]. Excess thyroid hormone increases cardiac Na+-K+ activated membrane ATPase, and sarcoplasmic reticulum Ca++-activated ATPase, both of which contribute to the heightened contractility of cardiac muscle. In addition excess thyroid hormone, at least in experimental animals, causes, by a direct effect on DNA transcription rates, an increased synthesis of alpha-myosin heavy chain with high ATPase activity, and decrease of beta myosin heavy chain synthesis. This alteration in alpha/beta ratio is associated with increased contractility [346].  In addition to the gene-mediated effects of thyroid hormone on the heart, triiodothyronine has direct effects that cause lower systemic vascular resistance and a higher cardiac output (347)

Whether an autoimmune process is involved in low output cardiac dysfunction in patients with Graves’ disease was investigated by myocardial biopsy of eleven patients in a study by Fatourechi and Edwards. Two of the group had lymphocytic infiltrates suggestive of an autoimmune process, whereas the others did not, indicating that this process may occur but would be an unusual cause of cardiac dysfunction [348].

Table 6. Cardiac Manifestation of Graves' Disease

•  Tachycardia

•  LVH and strain on EKG

•  Premature atrial and ventricular contractions

•  Atrial fibrillation

•  Congestive heart failure

•  Angina with (or without) coronary artery disease

•  Myocardial infarction

•  Systemic embolization

•  Death from cardiovascular collapse

•  Resistance to some drug effects (digoxin, coumadin)

•  Residual cardiomegaly

The cardiovascular changes may be due in part to increased sensitivity to circulating epinephrine. Thyroid hormone administration may increase, or not alter, the catecholamine content of the heart. Guanethidine partially restores cardiac dynamics to normal, perhaps by releasing catecholamine from the heart and thus reducing the cardiac stimulation caused by this agent. Guanethidine and beta-adrenergic blockers slow the tachycardia of thyrotoxicosis. Concomitant with this slowing is an increase in stroke volume, and there may be either a decrease [223] or little change in cardiac output [350]. T4 can increase the heart rate directly in a manner not mediated by catecholamines [351], and presumably this direct chromatotropic effect adds to coincident sympathetic effects on the heart. Thyrotoxicosis causes increased beta-adrenergic receptors in the heart and increased responsiveness to isoproterenol [352, 353].  Alpha-adrenergic and cholinergic receptors are reduced. In animal studies, adenyl cyclase may be activated by T3, but reduced cardiac levels of ATP limit the response through protein kinase activation. The net effect is beta-adrenergic sensitization and cholinergic desensitization. Thyrotoxicosis also increases beta-adrenergic receptors on a variety of tissues [355]. An increased number of -adrenergic receptors could cause hyper-responsiveness to adrenergic agonists, and could mediate the heightened plasma cAMP levels noted in thyrotoxic patients in the basal state (in some studies) and after assumption of an upright posture or administration of glucagon and epinephrine [356, 356]. Propranolol treatment normalizes the cAMP responses to these drugs and, of course, inhibits the action of beta-adrenergic agonists on the heart. Possibly these actions of propranolol explain its ability to reduce somewhat the consumption of oxygen in thyrotoxicosis.

The impact of the interrelation of excess thyroid hormone and the sympathetic nervous system in humans is not finally settled [357]. There is clear evidence for increased beta- adrenergic receptors in the heart and elsewhere in thyrotoxicosis, as inferred from animal studies. Responses of the thyrotoxic human to adrenergic agonists are probably not excessive in relation to responses in normal subjects, although this question is much debated. Beta-adrenergic blockade clearly reduces some pathophysiologic responses, including the increased nitrogen and oxygen consumption, toward but not to normal. Metabolism of the heart and other organs is stimulated directly by thyroid hormone, and sympathetic effects are additive. It remains possible that the sympathetic responses are in fact exaggerated; it is also clear that sympathetic responses do not "mediate" thyroid hormone action. Left ventricular reserve is impaired in thyrotoxicosis, and beta adrenergic blockage can lead to increased pulmonary artery pressure in some circumstances and further impair cardiac function (358). This suggests caution in administering beta-blockers to patients with severe hyperthyroidism and any evidence of circulatory dysfunction. We have seen administration of propranolol to patients with severe hyperthyroidism on rare instances to cause  cardiovacular collapse and shock.

While this discussion has concentrated on the cardiovascular effects of thyrotoxicosis, it is worth remembering that long-term mild excess of thyroid hormone causes impaired cardiac reserve and exercise capacity [358]. Subclinical thyrotoxicosis can alter cardiac function, with increased heart rate, increased left ventricular mass index, increased cardiac contractility, diastolic dysfunction, and induction of ectopic atrial beats or arrhythmias (359). Some of these changes are reversible when euthyroidism is restored.

Smit et al studied 25 patients with a history of differentiated thyroid carcinoma with more than 10 yr of TSH suppressive therapy with L-T4. Medication was titrated in a single-blinded fashion to establish continuation of TSH suppression (low-TSH group) or euthyroidism (euthyroid group). At baseline, diastolic function was impaired in all lopr-TSH patients as indicated by abnormal values for the peak flow of the early filling phase (E, 55.3 +/- 9.5 mm/sec), the ratio of E and the peak flow of the atrial filling phase (E/A ratio, 0.87 +/- 0.13), the early diastolic velocity obtained by tissue Doppler (E', 5.7 +/- 1.3 cm/sec), and the peak atrial filling velocity obtained by tissue Doppler (A', 6.8 +/- 1.4 cm/sec), prolonged E deceleration time (234 +/- 34 msec), and isovolumetric relaxation time (121 +/- 15 msec). After 6 months, significant improvements were observed in the euthyroid group in the E/A ratio (+41%; P < 0.001), E deceleration time (-18%; P = 0.006), isovolumetric relaxation time (-25%; P < 0.001), E' (+31%; P < 0.001), and the E'/A' ratio (+40%; P < 0.001). Prolonged subclinical hyperthyroidism is accompanied by diastolic dysfunction that is at least partly reversible after restoration of euthyroidism. Because isolated diastolic dysfunction may be associated with increased mortality, this finding is of clinical significance (359).

HEMATOLOGIC CHANGES

In most patients the hemoglobin and hematocrit are in the normal or low range [360]. Blood volume is increased, and the red cell mass is actually increased in some patients. In severe thyrotoxicosis normocytic anemia with hemoglobin concentrations as low as 8 - 9 g/dl may be observed. Hyperthyroid patients with anemia may show impaired iron use [361, 362]. Malnutrition may play a role in this decrease. These anemias are unresponsive to hematinic therapy, but the blood picture returns to normal when the thyrotoxicosis is controlled [262]. Iron deficiency or megaloblastic anemia is exceptional and requires a search for some explanation other than thyrotoxicosis. It is possible that thyrotoxicosis may increase the need for vitamin B12, as shown experimentally, and perhaps for folic acid. Also, there is an increased incidence of antigastric antibodies and mild pernicious anemia in patients with Graves' disease.The glucose-6-phosphate dehydrogenase activity of red cells is increased in thyrotoxicosis [363].

A relative lymphocytosis is frequently found in the peripheral blood due to neutropenia [364]. A relative and an absolute increase in the number of monocytes was noted years ago [365]. The monocyte count was between 10 and 15%; in only 2 of the 30 cases was it less than 10%. Relative lymphocytosis and relative monocytosis, with a normal or slightly low total white cell count, constitute the characteristic blood findings of Graves' disease. There is also an increase in the percentage and number of B lymphocytes and, as discussed previously, an altered ratio of T lymphocyte subsets. Significant pancytopenia with leukocyte counts under 3x109/l and neutrophiles under 2x109/l occasionally occurs, and if unrelated to drug therapy, tend to recover during treatment (366).

Graves' disease is often associated with mild thrombocytopenia, and occasionally with idiopathic thrombocytopenic purpura [367]. This co-occurrence is thought to reflect the autoimmune pathogenesis of both diseases. Fourteen percent of patients with ITP are reported to have coincident Graves' disease. Mild thrombocytopenia may disappear spontaneously or with treatment of hyperthyroidism, or if severe, may respond to glucocorticoid therapy [368]. Other more severe cases are managed as typical cases of ITP. Bone marrow examination may show normal or increased megakaryocytes [368]. Hyperthyroidism also induces a shortened platelet life span, believed to be due to more rapid clearing of normal platelets by an activated reticulo-endothelial system. Both anti-platelet antibodies and shortened platelet life span could contribute to the low or low-normal levels of platelets found in Graves' patients. It is reported that all patients with Graves' disease have evidence of IgG bound to platelets.

Coagulation is usually normal in spite of mild prolongation of the prothrombin time. Antihemophilic factor is often elevated in level and returns to normal with treatment. Hyperthyroidism can be associated with increased coagulability, in part through elevation of factor VIII. Cerebral venous thrombosis has been reported in association with thyrotoxicosis, suggesting that occasionally the propensity for coagulation can lead to serious consequences(369). Capillary fragility is increased. Severe liver damage caused by thyrotoxicosis and secondary congestive heart failure may be associated with a hemorrhagic tendency.

RETICULOENDOTHELIAL AND LYMPHATIC SYSTEMS

The reticuloendothelial and lymphocytic systems undergo hyperplasia. There may be generalized lymphadenopathy, and the thymus may be enlarged. Occasionally the thymus presents as an anterior mediastinal mass, but diminishes to normal size with control of the thyrotoxicosis [370]. Some authors have reported that the spleen tip can be felt in 20% of patients, but in our experience this finding is not common. The autoimmune responses in these patients have been detailed above. Several markers of augmented immune activity are elevated, including soluble CD30, a molecule released by T helper 2 cells, and IL-6 [371].

GASTROINTESTINAL FINDINGS

The appetite is characteristically increased. The effect of this increase is to offset, in part (sometimes completely), the loss of weight that might be expected from the increased catabolism. Indeed, the pattern of weight loss with increased appetite is nearly pathognomonic of thyrotoxicosis, although it may occur in diabetes mellitus and malabsorption or intestinal parasitism. A minority of patients complain of anorexia. Needless to say, they are likely to show the greatest weight loss. Nausea and vomiting are rare, but when they occur, they are serious. They are usually features of severe thyrotoxicosis but may also reflect hypercalcemia. In the presence of hypermetabolism, vomiting leads quickly to dehydration, ketosis, and perhaps avitaminosis.

The incidence of achlorhydria in exophthalmic goiter was found some years ago to be approximately 40% and slightly higher in myxedema [372]. The figures would be lower today. Berryhill and Williams [373]found that 73% showed a return of free hydrochloric acid after surgical thyroidectomy. Gastric enzyme production is decreased, and a mild chronic gastritis may be present [374]. Fasting serum gastrin levels, and responses to arginine, are increased [375].

Abdominal pain is an occasional symptom of thyrotoxicosis. Its nature and origin are obscure. Epigastric pain may suggest ulcer, gallbladder disease, or pancreatitis. Vomiting is sometimes associated with the pain.

The rate of absorption from the gastrointestinal tract is accelerated. The glucose tolerance curve may show an abnormally rapid rise and fall. Absorption of vitamin A is enhanced, and vitamin A formation from carotene is also increased.

Increased frequency of normal bowel movements is common, and occasionally diarrhea occurs. Transit time is decreased, and fat absorption may be impaired to the point of steatorrhea if fat intake is excessive [376].

The liver is frequently palpable in the absence of congestive heart failure, and is typically palpable with heart failure. Evidence of mild to severe liver disease may be found [377]. The plasma albumin level may be below 4 g/dl, and the globulin level above 3 g/dl. Galactose tolerance is impaired. There may be mild elevation of PT. The LFTs can give the impression of viral hepatitis. For example in one survey of 81 thyrotoxic patients, three-fourths had some LFT abnormality, including 31% with elevated bilirubin, 24% with elevated SGOT, 13% with elevated LDH, 26% with elevated SGPT, and 67% with elevated alkaline phosphatase (which of course may reflect bone metabolism). Cholesterol is often depressed. The abnormalities clear with treatment [378]. Bilirubin retention and jaundice are occasionally seen without evidence of congestive heart failure, but they are much more commonly found when this complication is also present. On hepatic biopsy the liver may be entirely normal histologically, even when there are abnormalities in chemical findings; alternatively, there may be evidence of focal collections of lymphocytes, decrease in glycogen, and occasionally death of cells. On electron microscopy, the mitochondria are increased in size and the smooth endoplasmic reticulum is hypertrophic. The glycogen level is decreased [379].The fine stellate scarrings seen in the livers of patients with severe thyrotoxicosis and reported in the earlier literature are rarely observed today. The cause of hepatic disease has been thought to be congestive heart failure, malnutrition, intercurrent infections, and a direct toxic effect of thyroid hormone. Malnutrition must play a role. Congestive heart failure by itself certainly can induce gross abnormalities in liver function, and presumably this insult is worsened by coincident thyrotoxicosis. The splanchnic blood flow is increased in thyrotoxicosis and the arteriovenous oxygen difference is greater than normal, but hepatic anoxia, at least in the portal areas, might occur even without circulatory failure if the metabolic demand for oxygen exceeds the supply.

Several patients who have been jaundiced without signs of congestive heart failure or other cause of hepatic dysfunction have been reported [380]. In two of these patients there was considerable elevation of the indirect-reacting bilirubin level. Studies in one patient showed that conjugation products of glucuronic acid were secreted into the urine in greatly increased quantities. This finding ruled out any absolute deficiency in the glucuronyl transferase enzyme in the liver. It was hypothesized that in certain thyrotoxic patients there is a great increase in metabolites that must be excreted via the glucuronyl transferase enzyme system. Since bilirubin competes relatively inefficiently in this enzyme system, it may be "crowded out" in the presence of an increased quantity of substrates. As a result, it may not be conjugated as rapidly as normally, and its concentration in the serum would therefore rise. All of these patients had residual abnormalities in bilirubin metabolism when euthyroid. This finding suggests that an underlying abnormality was present and was exacerbated by thyrotoxicosis. It is probable that the occasional thyrotoxic and jaundiced patient may actually suffer from an unrecognized separate abnormality, such as Gilbert's disease or posthepatitic liver dysfunction, brought to light by thyrotoxicosis.

URINARY TRACT

Polyuria and occasionally glycosuria are seen in uncomplicated thyrotoxicosis. Standard clinical renal function tests give normal results. Glycosuria may reflect accelerated absorption of sugar from the intestine and glucose intolerance.

In hyperthyroid animals and humans, the glomerular filtration rate and renal blood flow are on average increased, as are tubular transfer maxima for glucose and diodrast. The glomerular filtration rate and renal blood flow alterations probably are secondary to increased cardiac output, whereas the increased tubular activity may be a direct effect of thyroid hormone on renal function [381]. Polyuria does not indicate insensitivity to vasopressin, for the kidney responds normally to vasopressin with an increase in concentration of urine [382].

Hypercalcemia is a feature of severe thyrotoxicosis, but it rarely injures the kidneys. Occasionally hyposthenuria and uremia occur [307, 308], or more selective renal damage takes place. Huth et al [383] reported a patient with renal tubular acidosis coincident with hyperthyroidism. Circumstantial evidence suggested that hyperthyroidism led to hypercalcemia, which in turn had damaged the renal medulla and produced acidosis.

ENDOCRINE SYSTEM

( TABLE 10-7)

Table 7. Changes in Endocrine Function in Graves' Disease

 

•  FTI and T3 increased, TSH reduced

•  Prolactin normal

•  Growth hormone normal

•  Parathyroid hormone suppressed

•  Cortisol normal, urinary 17-OHCS increased, urinary free cortisol normal

•  Free testosterone reduced in males

•  Diabetic control worsened

 

FEMALE REPRODUCTIVE SYSTEM

Menstruation is characteristically decreased in volume. With severe thyrotoxicosis, the menstrual cycle may be either shortened or prolonged, and finally amenorrhea develops. The relative importance of a primary action of excess thyroid hormone on the ovary or uterus, and pituitary dysfunction, are unclear. In some cases, amenorrhea with a proliferative endometrium is found. This finding suggests failure of pituitary LH production and ovulation [384, 385].

Fertility is depressed, but pregnancy can develop. The incidence of miscarriage, premature delivery, pre-eclampsia and heart failure are increased by maternal hyperthyroidism [386, 387]. Evidence has been presented that high maternal thyroid hormone levels can lead to suppressed fetal TSH, lower fetal weight, and fetal death (388). Pregnancy, on the other hand, often ameliorates the symptoms of thyrotoxicosis due to Graves’ disease, but relapse is prone to occur in the 3-4 months following delivery (Figure 10-10). This topic is discussed in Chapter 14. Premature ovarian failure co-occurs with Graves' disease and thyroiditis in Multiple Endocrine Autoimmunity Type II [9]. Reduced fertility and increase miscarriage rates are associated with AITD and positive antibodies. Increased rates of thyroid dysfunction and positive antibody tests have been reported in infertile women (389). One study reports that treatment of euthyroid women with positive antibodies by administration of thyroxine reduced the incidence of miscarriage to the that found in anti body-negative women (390).

Figure 10. Clinical course of a patient who had transient exacerbation of Graves' disease on two occasions shortly following delivery.

Infants born to thyrotoxic mothers usually show no evidence of hyperthyroidism at birth. Fetal and neonatal thyrotoxicosis, fortunately infrequent events, are discussed in Chapter 14. Maternal thyrotoxicosis is associated with increased fetal loss (391), generally attributed to effects on the maternal system. However a recent study shows that elevated maternal thyroid hormone levels lead to elevated thyroid hormone levels in the fetus, and induce fetal loss. Surviving fetuses have lower birth weight (388). 

MALE REPRODUCTIVE SYSTEM

Gynecomastia, with ductal elongation and epithelial hyperplasia, occurs occasionally. [392] The circulating level of free estradiol may be elevated in these men [393-395]. Peripheral conversion of testosterone and androstenedione to estrone and estradiol is increased in both sexes during hyperthyroidism [261]. This elevation probably accounts in part for the abnormality. In addition, the slightly elevated LH in men with gynecomastia suggests hypothalamic insensitivity to feedback control and some peripheral unresponsiveness to LH [393]. An imbalance between testosterone and estrogen may be related to gynecomastia.

Kidd et al [397] found impotence in half of a small group of thyrotoxic men and sperm counts below 40 million in four of five tested. In these studies, the total testosterone level was elevated, but because the testosterone-estrogen binding globulin level was also high, the free testosterone level was reduced and the response to hCG was blunted. In thyrotoxicosis, mean sperm density is lower, and fewer sperm have normal morphology. Motility is lower in thyrotoxic males. The abnormalities normalize when the patients become euthyroid (398). Thus, both Leydig cell and spermatogenic abnormalities may be present. Abalovich et al ( 399) reported similar findings, and in addition noted a high incidence of sperm abnormalities. All of the abnormalities returned to normal after therapy of the thyrotoxicosis. Radioiodine therapy can cause transient reductions in both sperm count and motility but do not seem to cause permanent effects with ordinary doses used in treatment under 14 mCi, equivalent to around 500 MBq (398).

PRL probably plays no role in these reproductive abnormalities, since in hyperthyroidism, its release tends to be inhibited both at the hypothalamic and pituitary level [400]. Surprisingly, galactorrhea, in women with normo-prolactinemia, is reported to occur in increased frequency [264].

ADRENAL CORTEX

There are no obvious signs or symptoms of altered adrenal cortical function in thyrotoxicosis, but distinct changes have been detected. In thyrotoxicosis the adrenal cortex is often hyperplastic. Administered adrenal steroids disappear from the plasma at an accelerated rate [265]. Their metabolism by reduction of the steroid nucleus is accelerated, and conjugation of the reduced steroids is proportionally increased. Since plasma corticoid levels are normal and their rate of metabolism is increased, total daily metabolism and excretion of 17-ketosteroids and 17 hydroxy-corticoids are usually increased [385,403].

Along with the accelerated plasma cortisol clearance of thyrotoxicosis, the pathways of metabolism are also altered. For example, thyrotoxicosis is associated with a relatively increased excretion of 11-oxycorticoid metabolites [267]. The 11-oxy compounds are biologically inactive. Because of the negative feedback control from the pituitary, this preferential channeling of steroids into the 11-oxy derivatives could be partly responsible for increased steroid production. There is increased production of steroids by the adrenal gland in order to maintain a normal concentration of active steroids in the peripheral blood and in the tissues [405]. Secretion of adrenocorticotropic hormone (ACTH) by the pituitary is reported to be increased [406]. There are increases in secretory episodes during the day, but the fall to zero secretion after midnight is retained.

A reduced response to exogenous ACTH [407] indicates that adrenal reserve is reduced. In fact, it has been hypothesized that in severe thyrotoxicosis and in thyroid storm there may be an element of adrenal insufficiency. This contention has not been proved. There is no reason to believe that T4 opposes the peripheral action of adrenal steroids.

An increase in the 5-alpha metabolite of testosterone (androsterone) and a relative decrease in the 5-beta metabolite (etiocholanolone) are seen in the urine of thyrotoxic patients [408], but no comparable change in adrenal corticoid metabolism has been observed. These interesting biochemical alterations could have physiologic significance, for the ketosteroid 5-alpha metabolites, such as androsterone, are biologically active. In hypothyroidism the reverse change occurs, that is, an increase in the biologically inactive 5-beta metabolites such as etiocholanolone. Because administration of large amounts of androsterone depresses the level of serum lipids, Hellman and co-workers [408] have hypothesized that this change in steroid metabolism may be a way in which T4 (or its lack) affects lipid metabolism and produces a depression or elevation in serum cholesterol concentration.

OTHER METABOLIC ASPECTS

The basal oxygen consumption in thyrotoxicosis, as measured by the BMR, is elevated. The increase is above the level of metabolism that the person would have if he or she were not thyrotoxic. In extreme thyrotoxicosis, the BMR may be double the standard, that is, according to the usual mode of expression, + 100. In moderately severe thyrotoxicosis it may be from +30 to +60, and in mild thyrotoxicosis from +10 to +30.

In addition to the BMR, one should consider the total metabolism, that is the basal plus the increments occasioned by work, food, or emotion. An increased cost of muscular work in thyrotoxicosis was reported many years ago by Plummer and Boothby [409] and Briard et al. [410] among others. These studies, whose results have been disputed, suggested that the thyrotoxic subject has less efficient coupling of oxidation and energy use than either the normal subject or, for example, the hypermetabolic patient with leukemia. Some recent studies indicate that the increase in energy expenditure caused by work is not altered by thyrotoxicosis [411], and other studies support the original observations.

CARBOHYDRATE METABOLISM

Absorption of carbohydrate from the intestine is accelerated, as is its removal from the plasma. After a standard oral glucose load is given, the thyrotoxic patient characteristically has an early and rapid rise in blood glucose concentration in 30 - 60 minutes (possibly to more than 200 mg/dl) and a rapid fall, so that by two hours the concentration is normal. The early peak may be associated with glycosuria. Intravenous administration does not usually elevate the blood glucose level beyond the rise found in normal subjects.

Thyrotoxicosis increases the demand for insulin, perhaps by accelerating its metabolism. In addition, resistance to the action of insulin is present, since in nondiabetic thyrotoxic patients normal fasting blood glucose levels are associated with double the normal insulin concentration [412], and resistance is found on incubation of adipocytes in vitro [413]. Diabetes may be activated or intensified, and is ameliorated or may disappear when the thyrotoxicosis is treated. Long ago experimental diabetes was shown to result from long-standing thyrotoxicosis in the presence of partial pancreatectomy [414]. The adverse effect of hyperthyroidism on glucose control in patients with non-insulin dependent diabetes is caused by increased basal hepatic glucose production and reduced ability of insulin and glucose to suppress hepatic glucose production [415, 416]. Although hyperthyroidism increases the requirement for insulin, the effectiveness of exogenous insulin on carbohydrate metabolism is actually enhanced. If glucose is administered intravenously and continuously to a thyrotoxic subject so that blood sugar is maintained at a high level, administered insulin has an effect on glucose removal from the blood [416] that is greater than that in the normal subject.

Type I diabetes mellitus co-occurs with increased frequency in autoimmune thyroid disease. Post-partum thyroid dysfunction is especially common in patients with diabetes [417]. In recent years many similarities in immunologic phenomena have been noted in the two diseases, including insulin receptor antibodies and islet cell antibodies.

LIPIDS

The serum cholesterol level is depressed in thyrotoxicosis. There is an increase both in synthesis and in degradation, but the balance results in a new lower steady-state concentration in the serum. The cholesterol pool in the body is altered by thyroid hormone in different directions, depending on the species involved, and does not necessarily parallel the serum cholesterol level. Hypocholesterolemia may be produced without a distinct decrease in total body or liver cholesterol [418-428]. Part of the cholesterol-lowering action of thyroid hormone may be due to the effects of malnutrition and weight loss, and part may be simply a manifestation of hypermetabolism, since agents that elevate metabolism, such as salicylates, also lower serum lipid levels.

Thyroid hormone directly enhances conversion of cholesterol to bile acids and their excretion in the bile. This metabolic route accounts for the disposal of 70-90% of the cholesterol formed in the body [421]. Thyroid hormone may also affect cholesterol metabolism by directly increasing the number of membrane surface low-density lipoprotein (LDL) receptors [422]. The increased cholesterol synthesis, and even more enhanced clearance rate in hyperthyroidism has been confirmed in a recent study. Hepatic lypogenesis is also strikingly increased, probably both by direct action of thyroid hormones and an increase in insulin. Triglyceride levels tend to be slightly elevated (423).

Levels of the other serum lipid components are lowered [424-429]. Plasma triglycerides are in the low normal range, and the clearance rate of infused triglycerides may be elevated. Postheparin lipolytic activity [425,426] may be low or normal. Hyperthyroidism causes lower levels of apo(a), HDL, and ratio of total/HDL cholesterol [420,426]. Plasma leptin levels are normal in hyperthyroid patients (427). The free tocopherol level of the plasma changes in parallel with the cholesterol alterations in thyrotoxicosis [429].

Rich et al. [430]found the level of nonesterified fatty acids elevated in thyrotoxic patients. This response can be seen within six hours after administration of L-T3 to normal subjects, and might be related to the observation that ketosis occurs more readily in thyrotoxic patients than in normal subjects.

PROTEIN METABOLISM

Protein formation and destruction are both accelerated in hyperthyroidism. Nitrogen excretion is increased, and nitrogen balance may be normal or negative, depending on whether intake meets the demands of increased catabolism. Testosterone is able to exert its anabolic effects in thyrotoxicosis.

Lewallen et al [431] found that administration of thyroid hormone increases albumin synthesis and degradation in normal subjects, increases the fractional rate of degradation, and reduces the quantity of exchangeable albumin.

Thyroid hormone in vivo [432] or in vitro [433], as reviewed in Chapter 2, exerts its basic action through stimulation of transcription and mRNA translation. Thyroid hormone stimulates incorporation of amino acids into protein by liver microsomes. This action is at least partially explained by increased production of mRNA and by an increased transfer of soluble tRNA- bound amino acids into microsomal protein [434]. Clinically, a great excess of thyroid hormone appears to have the opposite effect. Crispell et al. found that protein synthesis may be depressed by feeding thyroid hormone to normal humans [432]. Catabolism of collagen is increased, [435]and urinary hypodroxyproline excretion is characteristically increased. Gluconeogenesis from alanine is stimulated by thyroid hormone [436]. Probably the elevated somatomedin levels found in thyrotoxicosis [296] contribute to augmented protein synthesis.

L-carnitine feeding has been shown to reverse some of the metabolic abnormalities in Graves’ disease. In a randomized double-blind study, L-carnitine, in a dose of 2-4 grams/day also reversed or prevented some of the adverse effects of excess thyroid hormone on bone mineralization. Presumably this occurs because hyperthyroidism has depleted body stores of carnitine (437).

cAMP Metabolism

Basal cAMP levels are on average elevated in serum of thyrotoxic humans , and there is hyperresponsiveness to stimuli such as epinephrine and glucagon [438]. Urinary cAMP is likewise increased [439]. Treatment with propranolol (in some studies) lowers basal cAMP toward, but not to, normal [440]. Adrenergic receptors are increased in number, and responses are enhanced as compared to the normal state (see Chapter 2).

Vitamin Metabolism

The absorption of vitamin A is increased and conversion of carotene to vitamin A is accelerated in thyrotoxicosis. The requirements of the body are likewise increased, and low blood concentrations of vitamin A may be found.

Requirements for thiamine and vitamin B6 are increased [441]. Lack of the B vitamins has been implicated as a cause of liver damage in thyrotoxicosis. It has been demonstrated that vitamin B12 requirements are increased in experimental thyrotoxicosis.

Radiosensitivity

Radiosensitivity of animal and human tumors may be enhanced by administration of L-T3 [442].

DEATH IN THYROTOXICOSIS

Given current therapeutic resources, death from thyrotoxi cosis or from its treatment should be rare. Thyroid storm, when it occurs, can be a lethal event. In a study from Scottish hospitals, 20 of 33 hyperthyroid patients who died had congestive heart failure, 6 bronchial pneumonia, and 6 embolism in various sites. Nineteen had atrial fibrillation. Eight were considered to have thyroid storm [443]. One might suspect that several causes of death might be identified in any particular case, and that rarely if ever would pure thyrotoxicosis be the only assignable cause. In two recent deaths of which we are aware, in patients with fulminant thyrotoxicosis, the immediate cause was sudden cardiovascular collapse, with shock and arrhythmias including ventricular fibrillation, which was resistant to all usual resuscitative efforts.

DIFFERENCES BETWEEN THE SEXES

Over the years, we have become impressed that thyrotoxic men, considered collectively, present some striking differences from thyrotoxic women in their response to the disease. For a given rise in metabolism, the symptoms of women tend to be more conspicuous than do those of men. Of course, there are frequent exceptions. Men with moderately severe thyrotoxicosis often have few or minimal symptoms, whereas many women with much milder thyrotoxicosis often present an impressive array of symptoms.

A statistical study of symptoms and signs, chiefly circulatory, in a series of 184 thyrotoxic patients (52 men, 132 women), using 233 patients with nontoxic nodular goiter as controls [444], the male patients were somewhat older than the females, and there were more severe cases among men than among women. Cardiac symptoms were more common in women than in men, even though the men were older and more often had a severe form of the disease. Palpitations and dyspnea are both more common and more severe in the female than in the male group.

DIFFERENCES ATTRIBUTABLE TO AGE

Certain differences in symptoms are attributable to age. Classic exophthalmic goiter is relatively more frequent among younger patients, but severe infiltrative exophthalmos is virtually unknown before mid-adolescence. The character of the symptomatic response may be differentiated in another way [444]. In older patients circulatory symptoms are more in evidence. In older patients, emotional instability may be less evident, or depression may occur, and the symptoms and signs are manifestly circulatory. In many older patients with Graves' disease, the thyroid is not readily palpable. Anorexia in this group is fairly frequent, as is constipation. Devastating personality and emotional changes often appear in the child or adolescent with Graves' disease. Graves’ disease in very young children is associated problems in growth and development, including craniosynostosis. Children with thyrotoxicosis are tall for their age, and their bone ages are advanced at presentation. They tend to achieve final heights higher than their predicted target heights finally (445).

A study by Segni et al suggests that permanent brain damage can occur as a result of the illness (446).

REFERENCES

  1. Graves RJ: Clinical lectures. London Med Surg J (pt 2):516, 1835.
  2. DeGroot LJ, Quintans J. The causes of autoimmune thyroid disease. Endocrine Rev 10:537-562, 1989.
  3. Marino M, Chiovato L, Friedlander JA, Latrofa F, Pinchera A, McCluskey RT.1999Serum antibodies against megalin (GP330) in patients with autoimmune thyroiditis.J Clin Endocrinol Metab. 84:2468-2474.
  4. Morris JC, Bergert ER, Bryant WP: Binding of immunoglobulin G from patients with autoimmune thyroid disease to rat sodium-iodide symporter peptides: evidence for the iodide transporter as an autoantigen. Thyroid 7:527, 1997.
  5. Kubota S, Gunji K, Stolarski C, Kennerdell JS, Wall J: Reevaluation of the prevalences of serum autoantibodies reactive with "64-kd eye muscle proteins" in patients with thyroid-associated ophthalmopathy. Thyroid 8:175, 1998.
  6. Mariotti S, Kaplan EL, Medof ME, DeGroot LJ: Circulating thyroid antigen antibody immune complexes. Proceedings of the Eighth International Thyroid Congress, Sydney, Australia, February 3-8, 1980.
  7. Irvine WJ, Davies SH, Teitelbaum S et al: The clinical and pathological significance of gastric parietal cell antibody. Ann NY Acad Sci 124:657, 1965.
  8. Jenkins RC, Weetman AP. Disease associations with autoimmune thyroid disease.Thyroid. 2002 Nov;12(11):977-88
  9. Muir A, Schatz DA, MacLaren NK: Polyglandular failure syndromes. p. 3013. In DeGroot LJ (ed): Endocrinology, 3rd Ed. WB Saunders Co, Philadelphia, 1994.
  10. Katakura M, Yamada T, Aizawa T et al: Presence of antideoxyribonucleic acid antibody in patients with hyperthyroidism of Graves’ disease. J Clin Endocrinol Metab 64:405, 1987.
  11. Mackay IR, Taft LI, Cowling DC: Lupoid hepatitis. Lancet 2:1323, 1956.
  12. Siddiqi A, Monson JP, Wood DF, Besser GM, Burrin JM. Serum cytokines in thyrotoxicosis. J Clin Endocrinol Metab 84:435-439, 1999.
  13. Sonnet E, Massart C, Gibassier J, Allannic H, Maugendre D. 1999 Longitudinal study of soluble intercellular adhesion molecule-1 (ICAM-1) in sera of patients with Graves’ disease. J Endocrinol Invest. 22:430-435.
  14. Paggi A, Caccavo D, Ferri GM, Di Prima MA, Amoroso A, Vaccaro F, Bonomo L, Afeltra A. Anti-cardiolipin antibodies in autoimmune thyroid diseases. Clin Endocrinol 40:329-333, 1994.
  15. Ochi Y, DeGroot LJ: Vitiligo in Graves' disease. Ann Intern Med 71:935, 1969.
  16. Welti H: Skin manifestations associated with severe forms of Basedow’s disease. Bulletin der Schweirzerischen Akademie der Medizinischen Wissenschaften 23:476-482, 1968.
  17. Amoroso A, Garzia P, Pasquarelli C, Sportelli G, Afeltra A: Hashimoto’s thyroiditis associated with urticaria and angio-oedema: disappearance of cutaneous and mucosal manifestations after thyroidectomy. J Clin Pathol 50:254-256, 1997.
  18. Marino M, Ricciardi R, Pinchera A, Barbesino G, Manetti L, Chiovato L, Braverman LE, Rossi B, Muratorio A, Mariotti S: Mild clinical expression of myasthenia gravis associated with autoimmune thyroid diseases. J Clin Endocrinol Metab 82:438-443, 1997.
  19. Marshall J: ITP and Graves’ disease. Ann Intern Med 67:411, 1967.
  20. White RG, Bass BH, Williams E: Lymphadenoid goiter and the syndrome of systemic lupus erythematosus. Lancet 1:368, 1961.
  21. Mori T, Kriss JP: Measurements by competitive binding radioassay of serum antimicrosomal and antithyroglobulin antibodies in Graves’ disease and other thyroid disorders. J Clin Endocrinol Metab 33:688, 1971.
  22. Wang P-W, Huang M-J, Liu R-T, Chen CD: Triiodothyronine autoantibodies in Graves' disease: Their changes after antithyroid therapy and relationship with the thyroglobulin antibodies. Acta Endocrinol (Copenh) 122:22-28, 1990.
  23. Nakamura S, Sakata S, Shima H, Komaki T, Kojima N, Kamikubo K, Yasuda K, Miura K: Thyroid hormone autoantibodies (THAA) in two cases of Graves' disease: Effects of antithyroid drugs, prednisolone, and subtotal thyroidectomy. Endocrinol Japon 33:751-759, 1986.
  24. Kasagi K, Kousaka T, Higuchi K, Iida Y, Misaki T, Alam MS, Miyamoto S, Yamabe H, Konishi J: Clinical significance of measurements of antithyroid antibodies in the diagnosis of Hashimoto’s thyroiditis: Comparison with histological findings. Thyroid 6:445, 1996.
  25. Amino N, Hagen SR, Yamada N, Refetoff S: Measurement of circulating thyroid microsomal antibodies by the tanned red cell-hemagglutination technique. Its usefulness in the diagnosis of autoimmune thyroid disease. Clin Endocrinol 5:115, 1976.
  26. Mullins RJ, Cohen SBA, Webb LMC, Chernajovsky Y, Dayan CM, Londei M, Feldmann M: Identification of thyroid stimulating hormone receptor-specific T cells in Graves’ disease thyroid using autoantigen-transfected Epstein-Barr Virus-transformed B cell lines. J Clin Invest 96:30-37, 1995.
  27. Weetman AP, Gunn C, Hall R, McGregor AM: Thyroid autoantigen-induced lymphocyte proliferation in Graves’ disease and Hashimoto’s thyroiditis. J Clin Lab Immunol 17:1, 1985.
  28. Fisfalen M-E, DeGroot LJ, Quintans J, Franklin WA, Soltani K: Microsomal antigen reactive lymphocyte lines and clones derived from thyroid tissue of patients with Graves’ disease. J Clin Endocrinol Metab 66:776-784, 1988.
  29. Soliman M, Kaplan E, Fisfalen M-E, Okamoto Y, DeGroot LJ: T cell reactivity to recombinant human thyrotropin receptor extracellular domain and thyroglobulin in patients with autoimmune and non-autoimmune thyroid diseases. J Clin Endocrinol Metab 80:206-213, 1995.
  30. Aoki N, DeGroot LJ: Lymphocyte blastogenic response to human thyroglobulin in Graves' disease, Hashimoto's thyroiditis, and metastatic cancer. Clin Exp Immunol 38:523, 1979.
  31. DeGroot LJ, Kawakami Y, Fisfalen M-E, Okamoto Y, Yanagawa T. T cell epitopes in TPO and TSH receptor. Proceedings of the International Hashimoto Symposium--80th Anniversary of Hashimoto's Disease, Fukuoka, Japan, December 2-5, 1992.
  32. Soliman M, Kaplan EL, Yanagawa T, Hidaka Y, Fisfalen M-E, DeGroot LJ: T-cells recognize multiple epitopes in the human thyrotropin receptor extracellular domain. J Clin Endocrinol Metab 80:905-914, 1995.
  33. Kula D, Bednarczuk T, Jurecka-Lubieniecka B, Polanska J, Hasse-Lazar K, Jarzab M, Steinhof-Radwanska K, Hejduk B, Zebracka J, Kurylowicz A, Bar-Andziak E, Stechly T, Pawlaczek A, Gubala E, Krawczyk A, Szpak-Ulczok S, Nauman J, Jarzab B Interaction of HLA-DRB1 alleles with CTLA-4 in the predisposition to Graves' disease: the impact of DRB1*07. Thyroid.2006 May;16(5):447-53.
  34. Tandon N, Freeman M, Weetman AP: T cell responses to synthetic thyroid peroxidase peptides in autoimmune thyroid disease. Clin Exp Immunol 86:56-60, 1991.
  35. Fisfalen M-E, Soliman M, Okamoto Y, Soltani K, DeGroot LJ: Proliferative responses of T-cells to thyroid antigens and synthetic thyroid peroxidase peptides in autoimmune thyroid disease. J Clin Endocrinol Metab 80:1597-1604, 1995.
  36. Ploth DW, Fitz A, Schnetzker D et al: Thyroglobulin-anti-thyroglobulin immune complex glomerulone-phritis complicating radioiodine therapy, Clin Immunol Immunopathol 9:327, 1978.
  37. Matsuura M, Kikkawa Y, Akashi K et al: Thyroid antigen-antibody nephritis: Possible involvement of fucosyl-GMI as the antigen. Endocrinol Japon 34:587, 1987.
  38. Chiovato L, Bassi P, Santini F et al: Antibodies producing complement-mediated thyroid cytotoxicity in patients with atrophic or goitrous autoimmune thyroiditis. J Clin Endocrinol Metab 77:1700, 1993.
  39. Tektonidou MG, Anapliotou M, Vlachoyiannopoulos P, Moutsopoulos HM. Presence of systemic autoimmune disorders in patients with autoimmune thyroid diseases.Ann Rheum Dis. 2004 Sep;63(9):1159-61.
  40. der Kinderen PJ, Houstra-Lanz M, Schwarz F: Exophthalmos- producing substance in human serum. J Clin Endocrinol Metab 20:712, 1960.
  41. Dobyns BM, Steelman SL: The thyroid-stimulating hormone of the anterior pituitary as distinct from exophthalmos producing substance. Endocrinology 52:705, 1953.
  42. Adams DD: The presence of an abnormal thyroid-stimulating hormone in the serum of some thyrotoxic patients. J Clin Endocrinol Metab 18:699, 1958.
  43. Adams DD, Fastier FN, Howie JB, Kennedy TH, Kilpatrick JA, Stewart RDH: Stimulation of the human thyroid by infusions of plasma containing LATS protector. J Clin Endocrinol Metab 39:826, 1974.
  44. Smith BR, Hall R: Binding of thyroid stimulators to thyroid membranes. FEBS Lett 42:301, 1974.
  45. Huber GK, Safirstein R, Neufeld D, Davies TF: Thyrotropin receptor autoantibodies induce human thyroid cell growth and c-fos activation. J Clin Endocrinol Metab 72:1142-1147, 1991.
  46. Cornelis S, Uttenweiler-Joseph S, Panneels V, Vassart G, Costagliola S. Purification and characterization of a soluble bioactive amino-terminal extracellular domain of the human thyrotropin receptor. Biochemistry 40:9860-9869, 2001.
  47. Rees Smith B, McLachlan SM, Furmaniak J: Autoantibodies to the thyrotropin receptor. Endocr Rev 9:106-121, 1988.
  48. Ludgate ME, Vassart G: The thyrotropin receptor as a model to illustrate receptor and receptor antibody diseases. Baillieres Clin Endocrinol Metab 9:95-113, 1995.
  49. Orgiazzi J, Williams DE, Chopra IJ, Solomon DH: Human thyroid adenyl cyclase-stimulating activity in immunoglobulin G of patients with Graves' disease. J Clin Endocrinol Metab 42:341, 1976.
  50. Evans C, Morgenthaler NG, Lee S, Llewellyn DH, Clifton-Bligh R, John R, Lazarus JH, Chatterjee VKK, Ludgate M. Development of a luminescent bioassay for thyroid stimulating antibodies. J Clin Endocrinol Metab 84:374, 1999.
  51. Di Cerbo A, Di Paola R, Menzaghi C, Filippis VD, Tahara K, Corda D, et al. 1999 Graves' immunoglobulins activate phospholipase A2 by recognizing specific epitopes on thyrotropin receptor. J Clin Endocrinol Metab 84:3283-3292.
  52. Drexhage HA, Bottazzo GF, Doniach D, Bitensky L, Chayen J: Evidence for thyroid growth-stimulating immunoglobulins in some goitrous thyroid diseases. Lancet 2:287, 1980
  53. Endo K, Kasagi K, Konishi J, Ikekubo K, Okuno T, Takeda Y, Mori T, Torizuka K: Detection and properties of TSH-binding inhibitor immunoglobulins in patients with Graves' disease and Hashimoto's thyroiditis. J Clin Endocrinol Metab 46:734, 1978.
  54. Irvine WJ, Lamberg B-A, Cullen DR, Gordin R: Primary hypothyroidism preceding thyrotoxicosis: A report of 2 cases and a review of the literature. J Clin Lab Immunol 2:349, 1979.
  55. Eckstein AK, Plicht M, Lax H, Neuhauser M, Mann K, Lederbogen S, Heckmann C, Esser J, Morgenthaler NG.Thyrotropin receptor autoantibodies are independent risk factors for Graves' ophthalmopathy and help to predict severity and outcome of the disease. J Clin Endocrinol Metab. 2006 Sep;91(9):3464-70.
  56. Zakarija M, McKenzie JM: Isoelectric focusing of thyroid-stimulating antibody of Graves' disease. Endocrinology 103:1469, 1978.
  57. Endo K, Amir SM, Ingbar SH: Development and evaluation of a method for the partial purification of immunoglobulins specific for Graves' disease. J Clin Endocrinol Metab 52:113, 1981.
  58. Misrahi, M; Milgrom, E. Cleavage and shedding of the TSH receptor. Europ J Endocrinol 137 599-602 1997.
  59. McKenzie JM: Further evidence for a thyroid activator in hyperthyroidism. J Clin Endocrinol Metab 20:380, 1960. .
  60. Takasu N, Oshiro C, Akamine H, Komiya I, Nagata A, Sato Y, Yoshimura H, Ito K: Thyroid-stimulating antibody and TSH-binding inhibitor immunoglobulin in 277 Graves’ patients and in 686 normal subjects. J Endocrinol Invest 20:452-461, 1997.
  61. Bolk JH, Elte JWF, Bussemaker JK, Haak A, Van Der Heide D: Thyroid-stimulating immunoglobulins do not cause non- autonomous, autonomous, or toxic multinodular goiters. Lancet 1:61, 1979.
  62. Kuzuya N, Chiu SC, Ikeda H, Uchimura H, Ito K, Nagataki S: Correlation between thyroid stimulators and 3,5,3'-triiodo- thyronine suppressibility in patients during treatment for hyperthyroidism with thionamide drugs: Comparison of assays by thyroid-stimulating and thyrotropin-displacing activities. J Clin Endocrinol Metab 48:706, 1979.
  63. Soliman M, Kaplan EL, Abdel-Latif A, Scherberg N, DeGroot LJ: Does thyroidectomy, RAI therapy, or antithyroid drug treatment alter reactivity of patients’ T cells to epitopes of thyrotropin receptor in autoimmune thyroid diseases? J Clin Endocrinol Metab 80:2312-2321, 1995.
  64. Fenzi G, Hashizume K, Roudebush CP, DeGroot LJ: Changes in thyroid-stimulating immunoglobulins during antithyroid therapy. J Clin Endocrinol Metab 48:572, 1979.
  65. Mukhtar ED, Smith BR, Pyle GA et al: Relation of thyroid-stimulating immunoglobulins to thyroid function and effect of surgery, radioiodine, and antithyroid drugs. Lancet 1:713, 1975.
  66. Tahara K, Ishikawa N, Yamamoto K, Hirai A, Ito K, Tamura Y, Yoshida S, Saito Y, Kohn LD: Epitopes for thyroid stimulating and blocking autoantibodies on the extracellular domain of the human thyrotropin receptor. Thyroid 7:867, 1997.
  67. Mullins RJ, Cohen SBA, Webb LMC, Chernajovsky Y, Dayan CM, Londei M, Feldmann M: Identification of thyroid stimulating hormone receptor-specific T cells in Graves’ disease thyroid using autoantigen-transfected Epstein-Barr virus-transformed B cell lines. Clin Invest 96:30-37, 1995.
  68. Fisfalen M-E, Soliman M, Okamoto Y, Soltani K, DeGroot LJ. Proliferative responses of T cells to thyroid antigens and synthetic TPO peptides in autoimmune thyroid disease. J Clin Endocrinol Metab 80:1597-1604, 1995.
  69. Fisfalen M-E, Palmer EM, van Seventer GA, Soltani K, Sawai Y, Kaplan E, Hidaka Y, Ober C, DeGroot LJ: TSH-R and TPO specific T cell clones and their cytokine profile in autoimmune thyroid disease. J Clin Endocrinol Metab 82:3655-3663, 1997.
  70. Fisfalen M-E, Palmer EM, Van Seventer GA, Soltani K, Sawai Y, Kaplan E, Hidaka Y, Ober C, DeGroot, LJ. Thyrotropin-receptor and thyroid peroxidase-specific T cell clones and their cytokind profile in autoimmune thyroid disease. J Clin Endocrinol Metab 82:3655-3663, 1997.
  71. Mullins RJ, Cohen SBA, Webb LMC, Chernajovsky Y, Dayan CM, Londei M, Feldmann M. Identification of thyroid stimulating hormone receptor-specific T cells in Graves’ disease thyroid using autoantigen-transfected Epstein-Barr virus-transformed B cell lines. J Clin Invest 96:30-37, 1995.
  72. Inaba H, Martin W, Ardito M, De Groot AS, De Groot LJ.The role of glutamic or aspartic acid in position four of the epitope binding motif and thyrotropin receptor-extracellular domain epitope selection in Graves' disease.J Clin Endocrinol Metab. 2010 Jun;95(6):2909-16
  73. Crisp M, Starkey KJ, Lane C, Ham J, Ludgate M. Adipogenesis in thyroid eye disease. Investigative Ophthalmol Visual Sci 41:3249-3255, 2000.
  74. Bell A, Gagnon A, Grunder L, Parikh SJ, Smith TJ, Sorisky A. Functional TSH receptor in human abdominal preadipocytes and orbital fibroblasts. Amer J Physiol - Cell Physiol. 279:C335-C340, 2000.
  75. Haraguchi K, Shimura H, Kawaguchi A, Ikeda M, Endo T, Onaya T. Effects of thyrotropin on the proliferation and differentiation of cultured rat preadipocytes. Thyroid 9:613, 1999.
  76. Hiromatsu Y, Yang D, Bednarczuk T, Miyake I, Nonaka K, Inoue Y. Cytokine profiles in eye muscle tissue and orbital fat tissue from patients with thyroid-associated ophthalmopathy. J Clin Endocrinol Metab 85:1194-1199, 2000.
  77. Wakelkamp IM, Gerding MN, Van Der Meer JW, Prummel MF, Wiersinga WM. Both Th1- and Th2-derived cytokines in serum are elevated in Graves' ophthalmopathy. Clin Exper Immunol 121:453-457, 2000.
  78. Nygaard B, Metcalfe RA, Phipps J, Weetman AP, Hegedus L. Graves' disease and thyroid associated ophthalmopathy triggered by 131-I treatment of non-toxic goiter. J Endocrinol Invest 22:481-485, 1999.
  79. Ludgate M. Animal models of Graves' disease. Europ J Endocrinol 142:1-8, 2000.
  80. Flynn JC, Rao PV, Gora M, Alsharabi G, Wei W, Giraldo AA, David CS, BangaJP, Kong YM. Graves' hyperthyroidism and thyroiditis in HLA-DRB1*0301 (DR3) transgenic mice after immunization with thyrotropin receptor DNA.Clin Exp Immunol. 2004 Jan;135(1):35-40.
  81. Graves' animal models of Graves' hyperthyroidism. Thyroid. 2007 Oct;17(10):981-8.
  82. Horie I, Abiru N, Saitoh O, Ichikawa T, Iwakura Y, Eguchi K, Nagayama Y. Distinct role of T helper Type 17 immune response for Graves' hyperthyroidism in mice with different genetic backgrounds. Autoimmunity. 2011 Mar;44(2):159-65
  83. Smith TJ, Hoa N. Immunoglobulins from patients with Graves' disease induce hyaluronan synthesis in their orbital fibroblasts through the self-antigen, insulin-like growth factor-I receptor. J Clin Endocrinol Metab. 2004 Oct;89(10):5076-80
  84. Douglas RS, Naik V, Hwang CJ, Afifiyan NF, Gianoukakis AG, Sand D, Kamat S, Smith TJ. J Immunol. 2008 Oct 15;181(8):5768-74. B cells from patients with Graves' disease aberrantly express the IGF-1 receptor: implications for disease pathogenesis.
  85. Schwiebert C, Morgenthaler NG, Köhrle J, Eckstein A, Schomburg L. Autoantibodies to the IGF1 receptor in Graves' orbitopathy. J Clin Endocrinol Metab. 2013 Feb;98(2):752-60
  86. Krieger CC, Neumann S, Place RF, Marcus-Samuels B, Gershengorn MC. Bidirectional TSH and IGF-1 Receptor Cross Talk Mediates Stimulation of Hyaluronan Secretion by Graves' Disease Immunoglobins. J Clin Endocrinol Metab. 2015 Mar;100(3):1071-7.
  87. van Steensel L, Hooijkaas H, Paridaens D, van den Bosch WA, Kuijpers RW, Drexhage HA, van Hagen PM, Dik WA.PDGF enhances orbital fibroblast responses to TSHR stimulating autoantibodies in Graves' ophthalmopathy patients.J Clin Endocrinol Metab. 2012 Jun;97(6):
  88. Hamada N, DeGroot LJ, Portmann L, Yamakawa J, Noh J, Okamoto Y, Ohno M, Ito K, Morii H. Thyroid microsomal antigen in Graves’ thyroid is not different from that in normal thyroid. Endocrinol Japon 38:471-478, 1991.
  89. Holm IA, Manson JE, Michels KB, Alexander EK, Willett WC, Utiger RD.Smoking and other lifestyle factors and the risk of Graves' hyperthyroidism.Arch Intern Med. 2005 Jul 25;165(14):1606-11
  90. Abbas AK, Lichtman AH, Pober JS: Cellular and molecular immunology. Philadelphia: WB Saunders, 1991.
  91. Strominger JL: Developmental biology of T cell receptors. Science 244:943-950, 1989.
  92. Spitzweg C, Joba W, Heufelder AE. Expression of thyroid-related genes in human thymus. Thyroid 9:133, 1999.52.11. .
  93. Sospedra M, Ferrer-Francesh X, Dominguez O, Juan M, Foz-Sala M, Pujol-Borrell R. Transcription of a broad range of self antigens in human thymus suggests a role for central mechanisms in tolerance toward peripheral antigens. J Immunol 161:5918-5929, 1998.
  94. Giménez-Barcons M, Casteràs A, Armengol Mdel P, Porta E, Correa PA, Marín A, Pujol-Borrell R, Colobran R. Autoimmune predisposition in Down syndrome may result from a partial central tolerance failure due to insufficient intrathymic expression of AIRE and peripheral antigens. J Immunol. 2014 Oct 15;193(8):3872-9
  95. Wenzel BE, Franke TF, Heufelder AE: Autoimmune thyroid diseases and enteropathogenic Yersinia enterocoliitica. Autoimmunity 7:295-303, 1990.
  96. Wolf MW, Misaki T, Bech K, Tvede M, Silva JE, Ingbar SH: Immunoglobulins of patients recovering from Yersinia enterocolitica infections exhibit Graves’ disease-like activity in human thyroid membranes. Thyroid 1:315-320, 1991.
  97. Wenzel BE, Heesemann J, Wenzel KW, Scriba PC: Patients with autoimmune thyroid diseases have antibodies to plasmid encoded proteins of enteropathogenic Yersinia. J Endocrinol Invest 11:139-140, 1988.
  98. Weiss M, Ingbar SH, Winblad S, Kasper DL: Demonstration of a saturable binding site for thyrotropin in Yersinia enterocolitica. Science 219:1331-1333, 1983.
  99. Werner J, Gelderblom H: Isolation of foamy virus from patients with de Quervain thyroiditis. Lancet 2:258-259, 1979.
  100. Nakachi K, Takasu N, Akamine H, Komiya I, Ishikawa K, Shinjyo T, Masuda M: Association of HLTV-1 with autoimmune thyroiditis in patients with adult T-cell leukemia (ATL) and in HTLV-1 carriers and a patient of ATL with autoimmune thyroiditis and uveites. Abstract No. PO27, presented at the 6th Asia and Oceania Thyroid Association Congress, November 9-12, 1997, Osaka, Japan.
  101. de Luis DA, Varela C, de la Calle H, Canton R, de Argila CM, San Roman AL, Boixeda D. Helicobacter pylori infection is markedly increased in patients with autoimmune atrophic thyroiditis. J Clin Gastroenterol 26:259-263, 1998.
  102. Szlachcic A, Sliwowski Z, Karczewska E, Bielanski W, Pytko-Polonczyk J, Konturek SJ. Helicobacter pylori and its eradication in rosacea. J Physiol Pharmacol 50:777-786. 1999.
  103. Rochman H, DeGroot LJ, Rieger CHL, Varnavides LA, Refetoff S, Joung JI, Hoye K: Carcinoembryonic antigen and humoral antibody response in patients with thyroid carcinoma. Cancer Research 35:2689-2692, 1975.
  104. Hancock SL, Cox RS, McDougall IR: Thyroid diseases after treatment of Hodgkin's disease. N Engl J Med 325:599, 1991.
  105. Vermiglio F, Castagna MG, Volnova E, Lo Presti VP, Moleti M, Violi MA, et al. 1999 Post-Chernobyl increased prevalence of humoral thyroid autoimmunity in children and adolescents from a moderately iodine-deficient area in Russia. Thyroid 9:781.
  106. Monzani F, Del Guerra P, Caraccio N, Casolaro A, Lippolis PV, Goletti O: Appearance of Graves’ disease after percutaneous ethanol injection for the treatment of hyperfunctioning thyroid adenoma. J Endocrinol Invest 20:294-298, 1997.
  107. Nygaard B, Helmer Knudsen J, Hegedus L, Vege Cand Scient A, Erik Molholm Hansen J: Thyrotropin receptor antibodies and Graves’ disease, a side effect of 131-I treatment in patients with nontoxic goiter. J Clin Endocrinol Metab 82:2926-2930, 1997.
  108. Schmidt M, Gorbauch E, Dietlein M, Faust M, Stutzer H, Eschner W, Theissen P, Schicha HIncidence of postradioiodine immunogenic hyperthyroidism/Graves' disease in relation to a temporary increase in thyrotropin receptor antibodies after radioiodine therapy for autonomous thyroid disease. Thyroid.2006 Mar;16(3):281-8.
  109. Klavinskis LS, Notkins AL, Oldstone MBA: Persistent viral infection of the thyroid gland: alteration of thyroid function in the absence of tissue injury. Endocrinology 122:567, 1988.
  110. Ronnblom LE, Alm GV, Oberg KE: Autoimmunity after alpha-interferon therapy for malignant carcinoid tumors. Ann Intern Med 115:178-183, 1991.
  111. Van Liessum PA, De Mulder PHM, Mattijssen EJM, Corstens FHM, Wagener DJT: Hypothyroidism and goiter during interleukin-2 therapy without LAK cells. Lancet 1:224, 1989.
  112. Hoekman K, von Blomberg-Van Der Flier BME, Wagstaff J, Drexhage HA, Pinedo HM: Reversible thyroid dysfunction during treatment with GM-CSF. Lancet 338:541-542, 1991.
  113. Brix TH, Christensen K, Holm NV, Harvald B, Hegedus L. A population-based study of Graves’ disease in Danish twins. Clinical Endocrinol 48:397-400, 1998
  114. Coles AJ, Wing MG, Hale G, Waldmann H, Weetman AP, Compston DAS, Chatterjee VKK. Anti-lymphocyte monoclonal antibody treatment of multiple sclerosis suppresses disease activity but induces Graves’ disease. J Endocrinol Invest 21:3, 1998
  115. Figueroa-Vega N, Alfonso-Pérez M, Benedicto I, Sánchez-Madrid F, González-Amaro R, Marazuela M. Increased circulating pro-inflammatory cytokines and Th17 lymphocytes in Hashimoto's thyroiditis. J Clin Endocrinol Metab. 2010 Feb;95(2):953-62
  116. Prummel, MF; Laurberg, P. Interferon- and autoimmune thyroid disease.Thyroid 13 547 2003
  117. Durelli L, Ferrero B, Oggero A, Verdun E, Ghezzi A, Montanari E, Zaffaroni M, the Betaferon Safety Trial Study Group. Thyroid function and autoimmuinity during Interferon alpha-1b treatment: A multicenter prospective study. J Clin Endocrinol Metab 86:3525-3532, 2001
  118. Bartels ED: Heredity in Graves' Disease. Copenhagen, Enjnar Munksgaards Forlag, 1941.
  119. Martin L: The hereditary and familial aspect of exophthalmic goiter and nodular goiter. Q J Med 14:207, 1945.
  120. Harvald B, Hauge M: A catamnestic investigation of Danish twins. Dan Med Bull 3:150, 1956.
  121. Brix TH, Kyvik KO, Christensen K, Hegedus L. Evidence for a major role of heredity in Graves’ disease: A population-based study of two Danish twin cohorts. J Clin Endocrinol Metab 86:930-934, 2001.
  122. Hansen PS, Brix TH, Iachine I, Kyvik KO, Hegedus L. The relative importance of genetic and environmental effects for the early stages of thyroid autoimmunity: a study of healthy Danish twins. Eur J Endocrinol.2006 Jan;154(1):29-38.
  123. Grumet FC, Payne RO, Konishi J, Kriss JP: HL-A antigens as markers for disease susceptibility and autoimmunity in Graves' disease. J Clin Endocrinol Metab 39:1115, 1974.
  124. Farid NR, Stone E, Johnson G: Graves' disease and HLA: Clinical and epidemiologic associations. Clin Endocrinol 13:535, 1980.
  125. Mangklabruks A, Cox N, DeGroot LJ: Genetic factors in autoimmune thyroid disease analyzed by restriction fragment length polymorphisms of candidate genes. J Clin Endocrinol Metab 73:236-244, 1991.
  126. Yanagawa T, Mangklabruks A, Chang Y-B, Okamoto Y, Fisfalen M-E, Curran PG, DeGroot LJ. Human histocompatibility leukocyte antigen-DQA1*0501 allele associated with genetic susceptibility to Graves' disease in a Caucasian population. J Clin Endocrinol Metab 76:1569-1574, 1993.
  127. Yanagawa T, Mangklabruks A, DeGroot LJ. Strong association between HLA-DQA1*0501 and Graves’ disease in a male Caucasian population. J Clin Endocrinol Metab 79:227-229, 1994.
  128. Chen Q-Y, Huang W, She J-X, Baxter F, Volpe R, MacLaren NK. 1999 HLA-DRB1*08, DRB1*03/DRB3*0101, and DRB3*0202 are susceptibility genes for Graves' disease in North American Caucasians, whereas DRB1*07 is protective. J Clin Endocrinol Metab 84:3182-3186.
  129. Trowsdale J, Ragoussis J, Campbell RD: Caption for Figure 2: The map of the human MHC is a compilation of physical mapping and cloning data from a number of laboratories. Immunol Today 14:349-352, 1993.
  130. Geluk A, Van Meijgaarden KE, Janson AAM, Wouter Drijfhout J, Meloen RH, DeVries RRP, Ottenhoff THM: Functional analysis of DR17(DR3)-restricted mycobacterial T cell epitopes reveals DR17-binding motif and enables the design of allele-specific competitor peptides. J Immunol 149:2864-2871, 1992.
  131. Sawai Y, DeGroot LJ. Binding of human thyrotropin receptor peptides to a Graves' disease predisposing human leukocyte antigen Class II molecule. J Clin Endocrinol Metab 85:1176-1179, 2000.
  132. Farid NR, Sampson L, Moens H, Barnard JM: The association of goitrous autoimmune thyroiditis with HLA-DR5. Tissue Antigens 21:173-175, 1981.
  133. Kong YC, Lomo LC, Motte RW, Giraldo AA, Baisch J, Strauss G, Hammerling GJ, David CS. HLA-DRB1 polymorphism determines susceptibility to autoimmune thyroiditis in transgenic mice: definitive association with HLA-DRB1*0301 (DR3) gene. J Exp Med 184:1167-1172, 1996.
  134. Pichurin, P; Chen, C-R; Pichurina, O; David, C; Rapoport, B; McLachlan, SM. Thyrotropin receptor-DNA vaccination of transgenic mice expressing HLA-DR3 or HLA-DQ6b. Thyroid 13 911-917 2003.
  135. Yanagawa T, Hidaka Y, Guimaraes V, Soliman M, DeGroot LJ: CTLA-4 gene polymorphism associated with Graves’ disease in a Caucasian population. J Clin Endocrinol Metab 80:41-45, 1995.
  136. Yanagawa T, Taniyama M, Enomoto S, Gomi K, Maruyama H, Ban Y, Saruta T: CTLA4 gene polymorphism confers susceptibility to Graves’ disease in Japanese. Thyroid 7:843, 1997.
  137. Thompson CB: Distinct roles for the costimulatory ligands B7-1 and B7-2 in T helper cell differentiation? Cell 81:979-982, 1995.
  138. Karandikar NJ, Vanderlugt CL, Walunas TL, Miller SD, Bluestone JA: CTLA4: A negative regulator of autoimmune disease. J Exp Med 184:783-788, 1996.
  139. Donner H, Rau H, Walfish PG, et al: CTLA4 alanine-17 confers genetic susceptibility to Graves’ disease and to Type I Diabetes Mellitus. J Clin Endocrinol Metab 82:143-146, 1997.
  140. Heward JM, Allahabadia A, Armitage M, Hattersley A, Dodson PM, MacLeod K, et al. 1999 The development of Graves' disease and the CTLA-4 gene on chromosome 2q33. J Clin Endocrinol Metab 84:2398-2401.
  141. Vaidya B, Imrie H, Perros P, Young ET, Kelly WF, Carr D, et al. 1999 The cytotoxic T lymphocyte antigen-4 is a major Graves' disease locus. Human Molecul Genet 8:1195-1199.
  142. Zaletel, K; Krhin, B; Gaberscek, S; Pirnat, E; Hojker, S. The influence of the exon 1 polymorphism of the cytotoxic T lymphocyte antigen 4 gene on thyroid antibody production in patients with newly diagnosed Graves’ disease. Thyroid 12 373 2002
  143. Kouki T, Sawai Y, Gardine C, Fisfalen M-E, Alegre M-L, DeGroot LJ. CTLA-4 gene polymorphism at position 49 in exon 1 reduces the inhibitory function of CTLA-4 and contributes to the pathogenesis of Graves' disease. J Immunol 165:6606-6611, 2000.
  144. Cuddihy RM, Dutton CM, Bahn RS: A polymorphism in the extracellular domain of the thyrotropin receptor is highly associated with autoimmune thyroid disease in females. Thyroid 5:89-95, 1995.
  145. Kotsa KD, Watson PF, Weetman AP: No association between a thyrotropin receptor gene polymorphism and Graves’ disease in the female population. Thyroid 7:31, 1997.
  146. Hiratani H, Bowden DW, Ikegami S, Shirasawa S, Shimizu A, Iwatani Y, Akamizu T Multiple SNPs in intron 7 of thyrotropin receptor are associated with Graves' disease. J Clin Endocrinol Metab. 2005 May;90(5):2898-903. Epub 2005 Mar 1.
  147. Dechairo BM, Zabaneh D, Collins J, Brand O, Dawson GJ, Green AP, Mackay I,Franklyn JA, Connell JM, Wass JA, Wiersinga WM, Hegedus L, Brix T, Robinson BG,Hunt PJ, Weetman AP, Carey AH, Gough SC.Association of the TSHR gene with Graves' disease: the first disease specific locus. Eur J Hum Genet. 2005 Aug 17;
  148. Vaidya B, Imrie H, Perros P, Young ET, Kelly WF, Carr D, Large DM, Toft AD, Kendall-Taylor P, Pearce SH. Evidence for a new Graves’ disease susceptibility locus at chromosome 18q21. Amer J Human Genetics 66:1710-1714, 2000.
  149. Ban Y, Taniyama M, Ban Y. Vitamin D receptor gene polymorphism is associated with Graves' disease in the Japanese population. J Clin Endocrinol Metab 85:4639-4643, 2000.
  150. Tomer Y, Concepcion E, Greenberg DA. A C/T single-nucleotide polymorphism in the region of the CD40 gene is associated with Graves’ disease. Thyroid 12:1129, 2002.
  151. Kurylowicz A, Kula D, Ploski R, Skorka A, Jurecka-Lubieniecka B, Zebracka J, Steinhof-Radwanska K, Hasse-Lazar K, Hiromatsu Y, Jarzab B, Bednarczuk T. Association of CD40 Gene Polymorphism (C-1T) with Susceptibility and Phenotype of Graves' Disease. Thyroid.2005 Oct;15(10):1119-24.
  152. Lopez ER, Zwermann O, Segni M, Meyer G, Reincke M, Seissler J, Herwig J, Usadel KH, Badenhoop K. A promoter polymorphism of the CYP27B1 gene is associated with Addison's disease, Hashimoto's thyroiditis, Graves' disease and type 1 diabetes mellitus in Germans. Eur J Endocrinol. 2004 Aug;151(2):193-7
  153. Shirasawa S, Harada H, Furugaki K, Akamizu T, Ishikawa N, Ito K, Ito K, Tamai H, Kuma K, Kubota S, Hiratani H, Tsuchiya T, Baba I, Ishikawa M, Tanaka M, Sakai K, Aoki M, Yamamoto K, Sasazuki T. SNPs in the promoter of a B cell-specific antisense transcript, SAS-ZFAT, determine susceptibility to autoimmune thyroid disease. Hum Mol Genet. 2004 Oct 1;13(19):2221-31. Epub 2004 Aug 04.
  154. Hiromatsu Y, Fukutani T, Ichimura M, Mukai T, Kaku H, Nakayama H, Miyake I, Shoji S, Koda Y, Bednarczuk T Interleukin-13 gene polymorphisms confer the susceptibility of Japanese populations to Graves' disease.. J Clin Endocrinol Metab. 2005 Jan;90(1):296-301.
  155. Ban Y, Tozaki T, Taniyama M, Tomita M, Ban Y. The Codon 620 Single Nucleotide Polymorphism of the Protein Tyrosine Phosphatase-22 Gene Does not Contribute to Autoimmune Thyroid Disease Susceptibility in the Japanese. Thyroid.2005 Oct;15(10):1115-8.
  156. Yamada H, Watanabe M, Nanba T, Akamizu T, Iwatani Y.The +869T/C polymorphism in the transforming growth factor-beta1 gene is associated with the severity and intractability of autoimmune thyroid disease.Clin Exp Immunol. 2008 Mar;151(3):379-82.
  157. Zeitlin AA, Simmonds MJ, Gough SC.Genetic developments in autoimmune thyroid disease: an evolutionary process.Clin Endocrinol (Oxf). 2007 Dec 13
  158. Sutherland A, Davies J, Owen CJ, Vaikkakara S, Walker C, Cheetham TD, James RA, Perros P, Donaldson PT, Cordell HJ, Quinton R, Pearce SH Genomic polymorphism at the interferon-induced helicase (IFIH1) locus contributes to Graves' disease susceptibility.J Clin Endocrinol Metab. 2007 Aug;92(8):3338-41.
  159. Liu N, Li X, Liu C, Zhao Y, Cui B, Ning G Hum Immunol. 2010 Feb 8 The association of interleukin-1alpha and interleukin-1beta polymorphisms with the risk of Graves' disease in a case-control study and meta-analysis.
  160. T aylor JC, Gough SC, Hunt PJ, Brix TH, Chatterjee K, Connell JM, Franklyn JA, Hegedus L, Robinson BG, Wiersinga WM, Wass JA, Zabaneh D, Mackay I, Weetman AP. A Genome-wide Screen in 1119 Relative Pairs with Autoimmune Thyroid Disease. J Clin Endocrinol Metab.2005 Nov 8;
  161. Berisso GA, van Lint MT, Bacigalupo A, Marmont AM. 1999 Adoptive autoimmune hyperthyroidism following allogeneic stem cell transplantation from an HLA-identical sibling with Graves' disease. Bone Marrow Transplantation. 23:1091-1092.
  162. Kisiel B, Bednarczuk T, Kostrzewa G, Kosińska J, Miśkiewicz P, Płazińska MT, Bar-Andziak E, Królicki L, Krajewski P, Płoski R.Polymorphism of the oestrogen receptor beta gene (ESR2) is associated with susceptibility to Graves' disease.Clin Endocrinol (Oxf). 2008 Mar;68(3):429-34
  163. Volpe R, Farid NR, Westarp CV, Row VV: The pathogenesis of Graves’ disease and Hashimoto’s thyroiditis. Clin Endocrinol (Oxf) 3:239, 1974.
  164. Lahl K, Loddenkemper C, Drouin C, Freyer J, Arnason J, Eberl G, Hamann A,Wagner H, Huehn J,Sparwasser T. Selective depletion of Foxp3+ regulatory T cells induces a scurfy-like disease.J Exp Med. 2007 Jan 22;204(1):57-63.
  165. Seddiki N, Santner-Nanan B, Martinson J, Zaunders J, Sasson S, Landay A,Solomon M, Selby W, Alexander SI, Nanan R, Kelleher A, Fazekas de St Groth B. Expression of interleukin (IL)-2 and IL-7 receptors discriminates between human regulatory and activated T cells.J Exp Med. 2006 Jul 10;203(7):1693-700.
  166. Banham AH, Powrie FM, Suri-Payer E. FOXP3+ regulatory T cells: Current controversies and future perspectives. Eur J Immunol. 2006 Nov;36(11):2832-6.
  167. Liu W, Putnam AL, Xu-Yu Z, Szot GL, Lee MR, Zhu S, Gottlieb PA, Kapranov P,Gingeras TR, Fazekas de St Groth B, Clayberger C, Soper DM, Ziegler SF,Bluestone JA. CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells. J Exp Med. 2006 Jul 10;203(7):1701-11.
  168. Raimondi G, Turner MS, Thomson AW, Morel PA. Naturally occurring regulatory T cells: recent insights in health and disease. Crit Rev Immunol. 2007;27(1):61-95.
  169. Beissert S, Schwarz A, Schwarz T. Regulatory T cells.J Invest Dermatol.2006 Jan;126(1):15-24.
  170. Pyzik M, Piccirillo CA TGF-{beta}1 modulates Foxp3 expression and regulatory activity in distinct CD4+ T cell subsets. J Leukoc Biol. 2007 May 2.
  171. Miyara M, Sakaguchi S. Natural regulatory T cells: mechanisms of suppression. Trends Mol Med. 2007 Mar;13(3):108-16.
  172. Pinkse GG, Tysma OH, Bergen CA, Kester MG, Ossendorp F, van Veelen PA,Keymeulen B, Pipeleers D, Drijfhout JW, Roep BO. Autoreactive CD8 T cells associated with beta cell destruction in type 1diabetes.Proc Natl Acad Sci U S A. 2005 Dec 20;102(51):18425-30.
  173. Alyanakian MA, You S, Damotte D, Gouarin C, Esling A, Garcia C, Havouis S, Chatenoud L, Bach JF.Diversity of regulatory CD4+T cells controlling distinct organ-specific autoimmune diseases. Proc Natl Acad Sci U S A. 2003 Dec 23;100(26):15806-11.
  174. You S, Leforban B, Garcia C, Bach JF, Bluestone JA, Chatenoud L. Adaptive TGF-beta-dependent regulatory T cells control autoimmune diabetes and are a privileged target of anti-CD3 antibody treatment. Proc Natl Acad Sci U S A. 2007 Apr 10;104(15):6335-40..PMID: 17389382
  175. You S, Leforban B, Garcia C, Bach JF, Bluestone JA, Chatenoud L. Adaptive TGF-beta-dependent regulatory T cells control autoimmune diabetes and are a privileged target of anti-CD3 antibody treatment. Proc Natl Acad Sci U S A. 2007 Apr 10;104(15):6335-40.
  176. Korn T, Reddy J, Gao W, Bettelli E, Awasthi A, Petersen TR, Bäckström BT, Sobel RA, Wucherpfennig KW, Strom TB, Oukka M, Kuchroo VK. Myelin-specific regulatory T cells accumulate in the CNS but fail to control autoimmune inflammation.Nat Med. 2007 Apr;13(4):423-31
  177. Alvarado-Sanchez B, Hernandez-Castro B, Portales-Perez D, Baranda L, Layseca-Espinosa E, Abud-Mendoza C, Cubillas-Tejeda AC, Gonzalez-Amaro R. Regulatory T cells in patients with systemic lupus erythematosus.J Autoimmun. 2006 Sep;27(2):110-8.
  178. Yu J, Heck S, Patel V, Levan J, Yu Y, Bussel JB, Yazdanbakhsh K Defective circulating CD25 regulatory T cells in patients with chronic immune thrombocytopenic purpura. Blood. 2008 Apr 17.
  179. Li X, Xiao BG, Xi JY, Lu CZ, Lu JH. Decrease of CD4(+)CD25(high)Foxp3(+) regulatory T cells and elevation of CD19(+)BAFF-R(+) B cells and soluble ICAM-1 in myasthenia gravis. Clin Immunol.2008 Feb;126(2):180-8.
  180. Nakano A, Watanabe M, Iida T, Kuroda S, Matsuzuka F, Miyauchi A, Iwatani Y. Apoptosis-induced Decrease of Intrathyroidal CD4(+)CD25(+) Regulatory T Cells.in Autoimmune Thyroid Diseases.Thyroid. 2007 Jan;17(1):25-31.
  181. Marazuela M, Garcia-Lopez MA, Figueroa-Vega N, de la Fuente H,Alvarado-Sanchez B, Monsivais- Urenda A, Sanchez-Madrid F, Gonzalez-Amaro R. Regulatory T cells in human autoimmune thyroid disease.J Clin Endocrinol Metab. 2006 Sep;91(9):3639-46. Epub 2006 Jun 27.
  182. Gangi E, Vasu C, Cheatem D, Prabhakar BS. IL-10-producing CD4+CD25+ regulatory T cells play a critical role in granulocyte-macrophage colony-stimulating factor-induced suppression of experimental autoimmune thyroiditis.J Immunol. 2005 Jun 1;174(11):7006-13.
  183. Molteni M, Rossetti C, Scrofani S, Bonara P, Scorza R, Kohn LD. Regulatory CD8+ T cells control thyrotropin receptor-specific CD4+ clones in healthy subjects.Cancer Detect Prev. 2003;27(3):167-74.
  184. Watanabe M, Yamamoto N, Maruoka H, Tamai H, Matsuzuka F, Miyauchi A, Iwatani Y. Independent involvement of CD8+ CD25+ cells and thyroid autoantibodies in disease severity of Hashimoto's disease.Thyroid. 2002 Sep;12(9):801-8.
  185. Vaidya B, Shenton BK, Stamp S, Miller M, Baister E, Andrews CD, Dickinson AJ, Perros P, Kendall-Taylor P. Thyroid. 2005 Sep;15(9):1073-8.Analysis of peripheral blood T-cell subsets in active thyroid-associated ophthalmopathy: absence of effect of octreotide-LAR on T-cell subsets in patients with thyroid-associated ophthalmopathy.
  186. Glick AB, Wodzinski A, Fu P, Levine AD, Wald DN. Impairment of regulatory T-cell function in autoimmune thyroid disease. Thyroid. 2013 Jul;23(7):871-8
  187. Klatka M, Grywalska E, Partyka M, Charytanowicz M, Kiszczak-Bochynska E, Rolinski J.Th17 and Treg cells in adolescents with Graves' disease. Impact of treatment with methimazole on these cell subsets. Autoimmunity. 2014 May;47(3):201-11
  188. Pan D, Shin YH, Gopalakrishnan G, Hennessey J, De Groot LJ: Regulatory T cells in Graves' disease. Clin Endocrinol (Oxf). 2009 Oct;71(4):587-93. Epub 2009 Feb 16.
  189. Mao C, Wang S, Xiao Y, Xu J, Jiang Q, Jin M, Jiang X, Guo H, Ning G, Zhang Y. Impairment of regulatory capacity of CD4+CD25+ regulatory T cells mediated by dendritic cell polarization and hyperthyroidism in Graves' disease.J Immunol. 2011 Apr 15;186(8):4734-43
  190. Pacini F, DeGroot LJ: Studies of immunoglobulin synthesis in cultures of peripheral T and B lymphocytes. Reduced T- suppressor cell activity in Graves' disease. Clin Endocrinol 18:219, 1983.
  191. Topliss DJ, Okita N, Lewis M, Row VV, Volpe R: Allosuppressor T lymphocytes abolish migration inhibition factor production in autoimmune thyroid disease: Evidence from radiosensitivity experiments. Clin Endocrinol 15:335, 1981.
  192. Sridama V, Pacini F, DeGroot LJ: Decreased suppressor T-lymphocytes in autoimmune thyroid diseases detected by monoclonal antibodies. J Clin Endocrinol Metab 54:316, 1982.
  193. Topliss D, How J, Lewis M, Row V, Volpe R: Evidence for cell-mediated immunity and specific suppressor T-lymphocyte dysfunction in Graves' disease and diabetes mellitus. J Clin Endocrinol Metab 57:700, 1983.
  194. Ando T, Imaizumi M, Graves PN, Unger P, Davies TF. Intrathyroidal fetal microchimerism in Graves’ disease. J Clin Endocrinol Metab 87:3315-3320, 2002.
  195. Bottazo GF, Pujol-Borrell R, Hanfusa T, Feldman F: Role of aberrant HLA-DR expression and antigen presentation in induction of endocrine autoimmunity. Lancet 2:1115, 1983.
  196. Matsunaga M, Eguchi K, Fukuda T, Kurata A, Tezuka H, Shimomura C, Otsubo T, Ishikawa N, Ito K, Nagataki S: Class II major histocompatibility complex antigen expression and cellular interactions in thyroid glands of Graves’ disease. J Clin Endocrinol Metab 62:723, 1986.
  197. Eguchi K, Otsubo T, Kawabe Y, Shimomura C, Ueki Y, Nakao H, Tezuka H, Matsunaga M, Fukuda T, Ishikawa N, Ito K, Nagataki S: Synergy in antigen presentation by thyroid epithelial cells and monocytes from patients with Graves’ disease. Clin Exp Immunol 72:84, 1988.
  198. Piccinini LA, MacKenzie WA, Platzer M, Davies TF: Lymphokine regulation of HLA-DR gene expression in human thyroid cell monolayers. J Clin Endocrinol Metab 64:543, 1987.
  199. Mukuta T, Arreaza G, Nishikawa M, Resetkova E, Jamieson C, Tamai H, Volpe R: Thyroid xenografts from patients with Graves’ disease in severe combined immunodeficient mice and NIH-beige-nude-xid mice. Clin Investigative Med 20:5-15, 1997.
  200. Leclere J, Bene MC, Duprez A, Faure G, Thomas JL, Vignaud JM, Burlet C: Behavior of thyroid tissue from patients with Graves’ disease in nude mice. J Clin Endocrinol Metab 59:175, 1984.
  201. Markmann J, Lo D, Naji A, Palmiter RD, Brinster RL, Heber Katz E: Antigen presenting function of class II MHC expressing pancreatic beta cells. Nature 336:476-479, 1988.
  202. Iversen K: Temporary rise in the frequency of thyrotoxicosis in Denmark 1941-1945. Copenhagen, Rosenkilde and Bagger, 1948.
  203. Chiovato L, Pinchera A: Stressful life events and Graves’ disease. Europ J Endocrinol 134:680-682, 1996.
  204. Radosavljevic VR, Jankovic SM, Marinkovic JM: Stressful life events in the pathogenesis of Graves’ disease. Europ J Endocrinol 134:699-701, 1996.
  205. Winsa B, Adami HO, Bergstrom R, Gamstedt A, Dahlberg PA, Adamson U, Jansson R, Karlsson FA: Stressful life events and Graves' disease. Lancet 338:1475-1479, 1991.
  206. Matos-Santos A, Nobre EL, Costa JG, Nogueira PJ, Macedo A, Galvao-Teles A, de Castro JJ. Relationship between the number and impact of stressful life events and the onset of Graves’ disease and toxic nodular goiter. Clin Endocrinol 55:15-19, 2001.
  207. Landsberg L: Catecholamines and hyperthyroidism. Clin Endocrinol Metab 6:697, 1977.
  208. Bruun E: Exophthalmic goiter developing after treatment with thyroid preparations. Acta Med Scand 122:13, 1945.
  209. Vagenakis AG, Wang CA, Bruger A, Maloof F, Braverman LE, Ingbar SH: Iodide-induced thyrotoxicosis in Boston. N Engl J Med 287:523, 1972.
  210. Stanbury JB, Ermans AE, Bourdoux P, Todd C, Oken E, Tonglet R, Vidor G, Braverman LE, Medeiros-Neto G: Iodine-induced hyperthyroidism: Occurrence and epidemiology. Thyroid 8:83-100, 1998.
  211. Vidor GI, Stewart JC, Wall JR, Wangel A, Hetzel BS: Pathogenesis of iodine-induced thyrotoxicosis: Studies in northern Tasmania. J Clin Endocrinol Metab 37:901, 1973. Nilsson G: Self-limiting episodes of jodbasedow. Acta Endocrinol 74:475, 1973.
  212. Stewart JC, Vidor GI, Butterfield IH, Hetzel BS: Epidemic thyrotoxicosis in northern Tasmania. Studies of clinical features and iodine nutrition. Aust NZ J Med 3:203, 1971.
  213. Boukis IA, Koutras DA, Souvantzoglou A, Evangelopolou A, Vrontakis A, Moulopoulos SD: Thyroid hormone and immunological studies in endemic goiter. J Clin Endocrinol Metab. 57:859-862, 1983.
  214. Rasooly L, Burek CL, Rose NR: Iodine-induced autoimmune thyroiditis in NOD-H-2h4 mice. Clin Immunol Immunopathol 81:287-292, 1996.
  215. Allen EM, Appel MC, Braverman LE: The effect of iodine ingestion on the development of spontaneous lymphocytic thyroiditis in the diabetes prone BB/W rat. Endocrinol 118:1977-1981, 1986.
  216. Plummer HS: Results of administering iodine to patients having exophthalmic goiter. J Amer Med Assn 80:1955, 1923.
  217. Feinberg WD, Hoffman DL, Owen CA: The effects of varying amounts of stable iodide on the function of the human thyroid. J Clin Endocrinol Metab 19:567, 1959.
  218. Greer MA, DeGroot LJ: The effect of stable iodide thyroid secretion in man. Metabolism 5:682, 1956. Ochi Y, Hachiya T, Yoshimura M, Shiomi K, Miyazaki T: Inhibitory effect of excess iodide on Graves' disease. Iodine Metab Thyroid Function 6:127, 1973.
  219. Buhler UK, DeGroot LJ: Effects of stable iodine on thyroid iodine release. J Clin Endocrinol Metab 29:1546, 1969.
  220. Schuppert F, Taniguchi S-I, Schroder S, Dralle H, Von Zur Muhlen A, Kohn LD: In vivo and in vitro evidence for iodide regulation of major histocompatibility complex class I and class II expression in Graves’ disease. J Clin Endocrinol Metab 81:3622-3628, 1996.
  221. Salvi M, Pedrazzoni M, Girasole G, Giuliani N, Minelli R, Wall JR, Roti E. Serum concentrations of proinflammatory cytokines in Graves' disease: effect of treatment, thyroid function, ophthalmopathy and cigarette smoking. Europ J Endocrinol 143:197-202, 2000.
  222. Wegelius O, Naumann J, Brunish R: Uptake of 35S labeled sulfate in the harderian and the ventral lachrymal glands of the guinea pig during stimulation with ophthalmotrophic pituitary agents. Acta Endocrinol 30:53, 1959.
  223. Lamberg BA, Ripatti J, Gordin A, Juustila H, Sivula A, Bjorkesten G: Chromophobe pituitary adenoma with acromegaly and TSH-induced hyperthyroidism associated with parathyroid adenoma. Acta Endocrinol 60:157, 1969.
  224. Weintraub BD, Gershengorn MC, Kourides IA, Fein H: Inappropriate secretion of thyroid-stimulating hormone. Ann Intern Med 95:339, 1981.
  225. Duprez L, Parma J, Van Sande J, Allgeier A, Leclere J, Schvartz C, Delisle M-J, Decoulx M, Orgiazzi J, Dumont J, Vassart G: Germline mutations in the thyrotropin receptor gene cause non-autoimmune autosomal dominant hyperthyroidism. Nature Genetics 7:396-401, 1994.
  226. Iversen K: Temporary rise in the frequency of thyrotoxicosis in Denmark 1941-1945. Copenhagen, Rosenkilde and Bagger, 1948.
  227. Kasagi K, Konishi J, Endo K, Mori T, Nagahara K, Makimoto K, Kuma K, Torizuka K: Adenylate cyclase activity in thyroid tissue from patients with untreated Graves' disease. J Clin Endocrinol Metab 51:492, 1980.
  228. Stanbury JB, Janssen MA: The iodinated albumin-like component of the plasma of thyrotoxic patients. J Clin Endocrinol Metab 22:978, 1962.
  229. Farran HEA, Shalom ES: Effect of L-tyrosine upon the protein bound iodine in thyrotoxicosis. J Clin Endocrinol Metab 26:918, 1966.
  230. Sterling K, Chodos RB: Radiothyroxine turnover studies in myxedema, thyrotoxicosis, and hypermetabolism without endocrine disease. J Clin Invest 35:806, 1956.
  231. Ingbar SH, Freinkel N: Studies on thyroid function and the peripheral metabolism of 131I-labeled thyroxine in patients with treated Graves' disease. J Clin Invest 37:1603, 1958.
  232. Uller RP, Van Herle AJ: Effect of therapy on serum thyroglobulin levels in patients with Graves' disease. J Clin Endocrinol Metab 46:747, 1978.
  233. Greer MA, Smith GE: Method for increasing the accuracy of the radioiodine uptake as a test for thyroid function by the use of desiccated thyroid. J Clin Endocrinol Metab 14:1374, 1954.
  234. Werner SC, Spooner M: A new and simple test for hyperthyroidism employing L-triiodothyronine and the twenty-four hour 131I uptake methods. Bull NY Acad Med 31:137, 1955.
  235. Clague R, Mukhtar ED, Pyle GA, Nutt J, Clark F, Scott M, Evered D, Smith BR, Hall R: Thyroid-stimulating immunoglobulins and the control of thyroid function. J Clin Endocrinol Metab 43:550, 1976.
  236. Lamberg BA, Ard A, Saarinen P, Totterman T, Makinen T: Response to TRH, serum thyroid hormone concentration, and serum markers of autoimmunity after antithyroid therapy in Graves' disease. J Endocrinol Invest 1:9, 1978.
  237. Chopra IJ, Chopra U, Orgiazzi J: Abnormalities of hypothalamo-hypophyseal-thyroid axis in patients with Graves' ophthalmopathy. J Clin Endocrinol Metab 37:955, 1973.
  238. Franco PS, Hershman JM, Haigler ED, Pittman JA Jr: Response to thyrotropin-releasing hormone compared with thyroid suppression tests in euthyroid Graves' disease. Metabolism 22:1357, 1973.
  239. Plummer HS: Results of administering iodine to patients having exophthalmic goiter. J Amer Med Assn 80:1955, 1923.
  240. Raben MS: The paradoxical effects of thiocyanate and of thyrotropin on the organic binding of iodine by the thyroid in the presence of large amounts of iodide. Endocrinology 45:296, 1949.
  241. Paris J, McConahey WM, Tauxe WN, Woolner LB, Bahn RC: The effect of iodides on Hashimoto's thyroiditis. J Clin Endocrinol Metab 21:1037, 1961.
  242. Suzuki H, Mashimo K: Significance of the iodide-perchlorate discharge test in patients with 131I-treated and untreated hyperthyroidism. J Clin Endocrinol Metab 34:332, 1972.
  243. Braverman LE, Woeber KA, Ingbar SH: Induction of myxedema by iodide in patients euthyroid after radioiodine or surgical treatment of diffuse toxic goiter. N Engl J Med 281:816, 1969.
  244. Ochi Y, Hachiya T, Yoshimura M, Shiomi K, Miyazaki T: Inhibitory effect of excess iodide on Graves' disease. Iodine Metab Thyroid Function 6:127, 1973.
  245. Buhler UK, DeGroot LJ: Effects of stable iodine on thyroid iodine release. J Clin Endocrinol Metab 29:1546, 1969.
  246. DeGroot L: Kinetic analysis of iodine metabolism. J Clin Endocrinol Metab 26:149, 1966.
  247. Furszyfer J, Kurland LT, McConahey WM, Elveback LR: Graves' disease in Olmsted County, Minnestoa, 1935 through 1967. Mayo Clin Proc 45:636, 1970.
  248. Tunbridge WMG, Evered DE, Hall R, Appleton D, Brewis M, Clark F, Grimley-Evans J, Young E, Bird T, Smith PA: The spectrum of thyroid disease in a community: The Wickham Survey. Clin Endocrinol 7:481, 1977.
  249. Vanderpump MPJ et al: Incidence of thyroid disorders in the community based on a twenty year follow-up of the Whickham survey population. Clinical Endocrinol 43:55-68, 1995.
  250. Hollowell JG, Staehling NW, Flanders WD, Hannon WH, Gunter EW, Spencer CA, Braverman LE. Serum TSH, T(4), and thyroid antibodies in the United States population (1988 to 1994): National Health and Nutrition Examination Survey (NHANES III). J Clin Endocrinol Metab. 2002 Feb;87(2):489-99
  251. Heimann P: Ultrastructure of human thyroid. Acta Endocrinol 53(suppl 110):5, 1966.
  252. Adams RD, Denny-Brown D, Pearson CM: In Diseases of Muscle. New York, Harper & Row Publishers Inc, 1962.
  253. Bostrom H, Hed R: Thyrotoxic myopathy and polymyositis in elderly patients: Differential-diagnostic viewpoints. Acta Med Scand 162:225, 1958.
  254. McEachern D, Ross WD: Chronic thyrotoxic myopathy: Report of three cases with review of previously reported cases. Brain 65:181, 1942.
  255. Askanazy M, Rutishauser E: Die Knochen der Basedow-Kranken: Beitrag zur latenten Osteodystrophia fibrosa. Virchows Arch 291:653, 1933.
  256. Dudgeon LS, Urquhart AL: Lymphorrhages in the muscles in exophthalmic goiter. Grain 49:182, 1926.
  257. Naffziger HC: Progressive exophthalmos after thyroidectomy. West J Surg Obstet Gynecol 40:530, 1932.
  258. Rundle FF, Pochin EE. Orbital tissues in thyrotoxicosis: Quantitative analysis relating to exophthalmos. Clin Sci 5:51, 1944.
  259. Terndrup TE, Heisig DG, Garceau JP. Sudden death associated with undiagnosed Graves' disease. J Emergency Med 8:553-555, 1990.
  260. Ezrin C, Swanson HE, Humphrey JG, Dawson JW, Hill FM: The cells of the human adenohypophysis in thyroid disorders. J Clin Endocrinol Metab 19:958, 1959.
  261. Scheithauer BW, Kovacs KT, Young Jr WF, Randall RV: The pituitary gland in hyperthyroidism. Mayo Clin Proc 67:22- 26, 1992.
  262. Beaver DC, Pemberton J de J: The pathologic anatomy of the liver in exophthalmic goiter. Ann Intern Med 7:687, 1933.
  263. Movitt ER, Gerstl B, Davis AE: Needle biopsy in thyrotoxicosis. Arch Intern Med 91:729, 1953.
  264. Piper J, Poulsen E: Liver biopsy in thyrotoxicosis. Acta Med Scand 127:439, 1947.
  265. Bergman TA, Mariash CN, Oppenheimer JH: Anterior mediastinal mass in a patient with Graves' disease. J Clin Endocrinol Metab 55:587-588, 1982.
  266. Fallon MD, Perry III HM, Bergfeld M, Droke D, Teitelbaum SL, Avioli LV: Exogenous hyperthyroidism with osteoporosis. Arch Intern Med 143:442-444, 1983.
  267. Follis RH: Skeletal changes associated with hyperthyroidism. Bull Johns Hopkins Hosp 92:405, 1953.
  268. Siddiqi A, Burrin JM, Noonan K, James I, Wood DF, Price CP, Monson JP. A longitudinal study of markers of bone turnover in Graves’ disease and their value in predicting bone mineral density. J Clin Endocrinol Metab 82:753-759, 1997.
  269. Shulz H, Low H, Ernster L, Sjostrand FS: Electronenmikroskopische Studien an Leberschnitten von Thyroxin-behandelten Ratten, in Sjostrand FS, Rhodin J (eds): European Regional Conference on Electron Microscopy, 1st Proceedings of the Stockholm Conference, Sept. 1956, New York, Academic Press Inc, 1957, p 134.
  270. White WH: On prognosis of secondary symptoms of exophthalmic goiter. Br Med J 2:151, 1886.
  271. Sattler H: In Marchand GW, Marchand JF (trans): Basedow's Disease. New York, Grune & Stratton Inc. 1952.
  272. Hegedus L, Hansen JM, Karstrup S: High incidence of normal thyroid gland volume in patients with Graves' disease. Clin Endocrinol 19:603-607, 1983.
  273. Werner S: Classification of the eye changes of Graves' disease. J Clin Endocrinol Metab 29:982, 1969.
  274. Leblond CP, Fertman MB, Puppel ID, Curtis GM: Radioiodine autography in studies of human goitrous thyroid glands. Arch Pathol Lab Med 41:510, 1946.
  275. Mahaux JE, Chamla-Soumenkoff J, Delcourt R, Levin S: Painful enlargement of left subtrapezoid lymph nodes in Graves' disease. Br Med J 1:384, 1971.
  276. Rumbyrt JS, Schocket AL. Chronic urticaria and thyroid disease.Immunol Allergy Clin North Am. 2004 May;24(2):215-23, vi. Review.
  277. Brownlie BE, Rae AM, Walshe JW, Wells JE. Psychoses associated with thyrotoxicosis - 'thyrotoxic psychosis.' A report of 18 cases, with statistical analysis of incidence. Europ J Endocrinol 142:438-444, 2000.
  278. Perrild H, Hansen JM, Arnung K, Olsen PZ, Danielsen U: Intellectual impairment after hyperthyroidism. Acta Endocrinol 112:185-191, 1986.
  279. Feibel JH, Campa JF: Thyrotoxic neuropathy (Basedow's paraplegia). J Neurol Neurosurg Psychiatry 39:491, 1976.
  280. Condon JV, Becka DR, Gibbs FA: Electroencephalographic abnormalities in hyperthyroidism. J Clin Endocrinol Metab 14:1511, 1954.
  281. Shaw PJ, Walls TJ, Newman PK, Cleland PG, Cartlidge NE. 1991 Hashimoto's encephalopathy: a steroid-responsive disorder associated with high antithyroid antibody titers-report of 5 cases. Neurology 41:228-233.
  282. Canton A, de Fabregas O, Tintore M, Mesa J, Codina A, Simo R. Encephalopathy associated to autoimmune thyroid disease: a more appropriate term for an underestimated condition? J Neurol Sci 176:65-69, 2000.
  283. Woodbury DM, Hurley RE, Lewis NG, McArthur MW, Copeland WW, Kirschrink JF, Goodman LS: Effect of thyroxine, thyroidectomy, and 6-N-propyl-2-thiouracil on brain function. J Pharmacol Exp Ther 106:331, 1952.
  284. Ravera JJ, Cervino JM, Fernandez G, Ferrari Foreade A, Malosetti H, Muxi F, MaggioloJ, Mussio Fournier JC, Rawak R: Two cases of Graves' disease with signs of a pyramidal lesion: Improvement in neurologic signs during treatment with antithyroid drugs. J Clin Endocrinol Metab 20:876, 1960.
  285. Swanson JW, Kelly, Jr. JJ, McConahey WM: Neurologic aspects of thyroid dysfunction. Mayo Clin Proc 56:504-512, 1981.
  286. Waldenstrom J: Acute thyrotoxic encephalomyopathy: Its cause and treatment. Acta Med Scand 121:251, 1945.
  287. Feibel JH, Campa JF: Thyrotoxic neuropathy (Basedow's paraplegia). J Neurol Neurosurg Psychiatry 39:491-497, 1976.173a. Bernal J. Action of thyroid hormone in brain. J Endocrinol Invest 25:268-288, 2002.
  288. Sensenbach W, Madison L, Eisenberg S, Ochs L: The cerebral circulation and metabolism in hyperthyroidism and myxedema. J Clin Invest 33:1434, 1954.174a.
  289. Whitfield AGW, Hudson WA: Chronic thyrotoxic myopathy. Q J Med 30:257, 1961.
  290. Gold HK, Spann JF Jr, Braunwald E: Effects of alterations in the thyroid state on the intrinsic contractile properties of isolated rat skeletal muscle. J Clin Invest 49:849, 1970.
  291. Sanderson KV, Adey WR: Electromyographic and endocrine studies in chronic thyrotoxic myopathy. J Neurol Neurosurg Psychiatry 15:200, 1952.
  292. Zurcher RM, Horber FF, Grunig BE, Frey FJ: Effect of thyroid dysfunction on thigh muscle efficiency. J Clin Endocrinol Metab 69:1082, 1989.
  293. Erkintalo M, Bendahan D, Mattei J-P, Fabreguettes C, Vague P, Cozzone PJ. Reduced metabolic efficiency of skeletal muscle energetics in hyperthyroid patients evidenced quantitatively by in vivo phosphorus-31 magnetic resonance spectroscopy. Metabolism 47:769-776, 1998.
  294. Fitch CD, Coker R, Dinning JS: Metabolism of creatine I-14C by Vitamin E deficient and hyperthyroid rats. Am J Physiol 198:1232, 1960.
  295. Thorn G: Creatine studies in thyroid disease. Endocrinology 20:628, 1936.
  296. Drachman DB: Myasthenia gravis and the thyroid gland. N Engl J Med 266:330, 1962.
  297. Engel AG: Thyroid function and periodic paralysis. Am J Med 30:327, 1961.
  298. Fisher J: Thyrotoxic periodic paralysis with ventricular fibrillation. Arch Intern Med 142:1362-1364, 1982.
  299. Fraser SA, Anderson JB, Smith DA, Wilson GM: Osteoporosis and fractures following thyrotoxicosis. Lancet 1:981, 1971.
  300. Rosen CJ, Adler RA. Longitudinal changes in lumbar bone density among thyrotoxic patients after attainment of euthyroidism. J Clin Endocrinol Metab 75:1531-1534, 1992.
  301. Langdahl BL, Loft AGR, Eriksen EF, Mosekilde L, Charles P. Bone mass, bone turnover, body composition, and calcium homeostasis in former hyperthyroid patients treated by combined medical therapy. Thyroid 6:161, 1996.
  302. Vestergaard, P; Mosekilde, L. Hyperthyroidism, bone mineral, and fracture risk – a meta-analysis. Thyroid 13 585 2003
  303. Jodar E, Martinez-Diaz-Guerra G, Azriel S, Hawkins F. Bone mineral density in male patients with L-thyroxine suppressive therapy and Graves’ disease. Calcified Tissue International 69:84-87, 2001.
  304. Uzzan B, Campos J, Cucherat M, Nony P, Boissel JP, Perret GY. Effects on bone mass of long term treatment with thyroid hormones: A meta-analysis. J Clin Endocrinol Metab 81:4278-4289, 1996.
  305. Krane SM, Brownell GL, Stanbury JB, Corrigan H: The effect of thyroid disease on calcium metabolism in man. J Clin Invest 35:874, 1956.
  306. Harvey RD, McHardy KC, Reid IW, Paterson F, Bewsher PD, Duncan A, Robins P: Measurement of bone collagen degradation in hyperthyroidism and during thyroxine replacement therapy using pyridinium cross-links as specific urinary markers. J Clin Endocrinol Metab 72:1189-1194, 1991.
  307. Sataline LR, Powell C, Hamwi GJ: Suppression of the hypercalcemia of thyrotoxicosis by corticosteroids. N Engl J Med 267:646, 1962.
  308. Sallin O: Hypercalcemic nephropathy in thyrotoxicosis. Acta Endocrinol 29:425, 1958.
  309. Epstein FH, Freedman LR, Levitin H: Hypercalcemia, nephrocalcinosis and reversible renal insufficiency associated with hyperthyroidism. N Engl J Med 258:782, 1958.
  310. Bortz W, Eisenberg E, Bowers CY, Pout M: Differentiation between thyroid and parathyroid causes of hypercalcemia. Ann Intern Med 54:610, 1961.
  311. David NJ, Verner JV, Engel FL: Diagnostic spectrum of hypercalcemia: Case reports and discussion. Am J Med 33:88, 1962.
  312. Kleeman CR, Tuttle S, Bassett SH: Metabolic observations in a case of thyrotoxicosis with hypercalcemia. J Clin Endocrinol Metab 18:477, 1958.
  313. Lukert BP, Higgins JC, Stoskopf MM: Serum osteocalcin is increased in patients with hyperthyroidism and decreased in patients receiving glucocorticoids. J Clin Endocrinol Metab 62:1056, 1986.
  314. Cooper DS, Kaplan MM, Ridgway EC, Maloof F, Daniels GH: Alkaline phosphatase isoenzyme patterns in hyperthyroidism. Ann Intern Med 90:164, 1979.
  315. Garrel DR, Delmas PD, Malaval L, Tourniaire J: Serum bone gla protein: A marker of bone turnover in hyperthyroidism. J Clin Endocrinol Metab 62:1052, 1986.
  316. Bouillon R, DeMoor P: Parathyroid function in patients with hyper- or hypothyroidism. J Clin Endocrinol Metab 38:999, 1974.
  317. Bouillon R, Muls E, DeMoor P: Influence of thyroid function on the serum concentration of 1,25-dihydroxyvitamin D3. J Clin Endocrinol Metab 51:793, 1980.
  318. Peerenboom H, Keck E, Kruskemper HL, Strohmeyer G: The defect of intestinal calcium transport in hyperthyroidism and its response to therapy. J Clin Endocrinol Metab 59:936, 1984.
  319. Rude RK, Oldham SB, Singer FR, Nicoloff JT: Treatment of thyrotoxic hypercalcemia with propranolol. N Engl J Med 294:431, 1976.
  320. Rosen HN, Moses AC, Gundberg C, Kung VT, Seyedin SM, Chen T, Holick M, Greenspan SL. Therapy with parenteral pamidronate prevents thyroid hormone-induced bone turnover in humans. J Clin Endocrinol Metab 77:664-669, 1993.
  321. Jodar E, Munoz-Torres M, Escobar-Jimenez F, Quesada M, Luna JD, Olea N. Antiresorptive therapy in hyperthyroid patients: longitudinal changes in bone and mineral metabolism. J Clin Endocrinol Metab 82:1989-1994, 1997.
  322. Barsotti MM, Targovnik JH, Verso TA: Thyrotoxicosis, hypercalcemia, and secondary hyperparathyroidism. Arch Intern Med 139:661, 1979
  323. Siafakas NM, Milona I, Salesiotou V, Filaditaki V, Tzanakis N, Bouros D. Respiratory muscle strength in hyperthyroidism before and after treatment. Amer Rev Respiratory Dis 146:1025-1029, 1992.
  324. Siafakas NM, Milona I, Salesiotou V, Filaditaki V, Tzanakis N, Bouros D. Respiratory muscle strength in hyperthyroidism before and after treatment. Amer Rev Respiratory Dis 146:1025-1029, 1992.
  325. Massey DG, Becklake MR, McKenzie JM, Bates DV: Circulatory and ventilatory response to exercise in thyrotoxicosis. N Engl J Med 276:1104, 1967.
  326. Stein M, Kinbel P, Johnson RL: Pulmonary function in hyperthyroidism. J Clin Invest 40:348, 1961.
  327. Hamolsky MW: Asthma and hyperthyroidism. J Allergy Clin Immunol 49:348, 1972.
  328. Reynolds J, Woody HB: Thyrotoxic mitral regurgitation. Am J Dis Child 122:544, 1971.
  329. Auer J, Scheibner P, Mische T, Langsteger W, Eber O, Eber B. Subclinical hyperthyroidism as a risk factor for atrial fibrillation. American Heart Journal 142:838-842, 2001.
  330. Sandler G, Wilson GM: The nature and prognosis of heart disease in thyrotoxicosis. Q J Med 28:347, 1959.
  331. Graettinger JS, Muenster JJ, Selverstone LA, Campbell JA: A correlation of clinical and hemodynamic studies in patients with hyperthyroidism with and without congestive heart failure. J Clin Invest 38:1316, 1959.
  332. Blizzard JJ, Rupp JJ: Prolongation of the P-R interval as a manifestation of thyrotoxicosis. J Amer Med Assn 173:1845, 1960.
  333. Resnekov L, Falicow R: Thyrotoxicosis and lactate-producing angina pectoris with normal coronary arteries. Brit Heart J 39:1051, 1977.
  334. Marti V, Ballester M, Rigla M, Narula J, Berna L, Pons-Llado G, Carrio I, Carreras F, Webb SM. Myocardial damage does not occur in untreated hyperthyroidism unless associated with congestive heart failure. Amer Heart J 134:1133-1137, 1997.
  335. Choi YH, Chung JH, Bae SW, Lee WH, Jeong EM, Kang MG, Kim BJ, Kim KW, Park JE .Severe coronary artery spasm can be associated with hyperthyroidism. Coron Artery Dis. 2005 May;16(3):135-9.
  336. Kotler N, Kyriakos M, Bouchard J, Warbasse JR: Myocardial infarction associated with thyrotoxicosis. Arch Intern Med 132:723, 1973.
  337. Rowe GG, Huston JH, Weinstein AB, Tuchman H, Brown JF, Crumpton CW: The hemodynamics of thyrotoxicosis in man with special reference to coronary blood flow and myocardial oxygen metabolism. J Clin Invest 35:272, 1956.
  338. Rodbard D, Fugita T, Rodbard S: Estimation of thyroid function by timing the arterial sounds. J Amer Med Assn 201:884, 1967.
  339. Buccino RA, Spann JF Jr, Sonnenblick EH, Braunwald E: Effect of thyroid state on myocardial contractility. Endocrinology 82:191, 1968.
  340. Osman F, Gammage MD, Sheppard MC, Franklyn JA. Cardiac dysrhythmias and thyroid dysfunction: the hidden menace? J Clin Endocrinol Metab 87:963-967, 2002.
  341. Woeber KA. Thyrotoxicosis and the heart. N Engl J Med 327:94-98, 1992.
  342. Kahaly GJ, Nieswandt J, Mohr-Kahaly S. Cardiac risks of hyperthyroidism in the elderly. Thyroid 8:1165, 1998.
  343. Mintz G, Pizzarello R, Klein I: Enhanced left ventricular diastolic function in hyperthyroidism: Noninvasive assessment and response to treatment. J Clin Endocrinol Metab 73:146-150, 1991.
  344. Valcavi R, Menozzi C, Roti E, Zini M, Lolli G, Roti S, Guiducci U, Portioli I. Sinus node function in hyperthyroid patients. J Clin Endocrinol Metab 75:239-242, 1992.
  345. Smolenski RT, Yacoub MH, Seymour AM. Hyperthyroidism increases adenosine transport and metabolism in the rat heart. Molecul Cellul Biochem 143:143-149, 1995.
  346. Dillmann WH: Biochemical basis of thyroid hormone action in the heart. Amer J Med 88:626-630, 1990
  347. Schmidt BMW, Martin N, Georgens AC, Tillmann H-C, Feuring M, Christ M, Wehling M. Nongenomic cardiovascular effects of triiodothyronine in euthyroid male volunteers. J Clin Endocrinol Metab 87:1681-1686, 2002. .
  348. Fatourechi V, Edwards WD. Graves' disease and low-output cardiac dysfunction: Implications for autoimmune disease in endomyocardial biopsy tissue from eleven patients. Thyroid 10:601, 2000.
  349. Pietras RJ, Real MA, Poticha GS, Bronsky D, Waldstein SS: Cardiovascular response in hyperthyroidism. The influence of adrenergic-receptor blockade. Arch Intern Med 129:426, 1972.
  350. deGroot WJ, Leonard JJ, Paley HW, Johnson JE, Warren JV: The importance of autonomic integrity in maintaining the hyperkinetic circulating dynamics of human hyperthyroidism. J Clin Invest 40:1033, 1961.
  351. McDevitt DG, Shanks RG, Hadden DR, Montgomery DAD, Weaver JA: The role of the thyroid in the control of the heart-rate. Lancet 1:997, 1968.
  352. Tse J, Wrenn RW, Kuo JF: Thyroxine-induced changes in characteristics and activities of beta-adrenergic receptors and adenosine 3',5'-monophosphate and guanosine 3',5'-monophosphate systems in the heart may be related to reputed catecholamine supersensitivity in hyperthyroidism. Endocrinology 107:6, 1980.
  353. Williams LT, Lefkowitz RJ, Watanabe AM, Hathaway DR, Besch H Jr: Thyroid hormone regulation of beta-adrenergic receptor number. J Biol Chem 252:2787, 1977.
  354. Ginsberg AM, Clutter WE, Shah SD, Cryer PE: Triiodothyro- nine-induced thyrotoxicosis increases mononuclear leukocyte beta-adrenergic receptor density in man. J Clin Invest 67:1785, 1981.
  355. Nilsson OR, Anderson RGG, Karlberg BE: Effects fo propranolol and atenolol on plasma and urinary cyclic adenosine 3',5'-monophosphate in hyperthyroid patients. Acta Endocrinol 94:38, 1980.
  356. Guttler RB, Croxon MS, De Quattro VL, Warren DW, Otis CL, Nicoloff JT: Effects of thyroid hormone on plasma adenosine 3',5'-monophosphate productin in man. Metabolism 26:1155, 1977.
  357. Levey GS, Klein I: Catecholamine-thyroid hormone interactions and the cardiovascular manifestations of hyperthyroidism. Amer J Med 88:642-646, 1990.
  358. Biondi B, Fazio S, Cuocolo A, Sabatini D, Nicolai E, Lombardi G, Salvatore M, Sacca L. Impaired cardiac reserve and exercise capacity in patients receiving long-term thyrotropin suppressive therapy with levothyroxine. J Clin Endocrinol Metab 81:4224-4228, 1996
  359. Smit JW, Eustatia-Rutten CF, Corssmit EP, Pereira AM, Frolich M, Bleeker GB, Holman ER, van der Wall EE, Romijn JA, Bax JJ. Reversible diastolic dysfunction after long-term exogenous subclinical hyperthyroidism: a randomized, placebo-controlled study. J Clin Endocrinol Metab. 2005 Nov;90(11):6041-7
  360. Nightingale S, Vitek PJ, Himsworth RL: The hematology of hyperthyroidism. Q J Med 47:35, 1978.
  361. Popovic WJ, Brown JE, Adamson JW: The influence of thyroid hormones on in vitro erythropoiesis. J Clin Invest 60:907, 1977.
  362. Rivlin RS, Wagner HN Jr: Anemia in hyperthyroidism. Ann Intern Med 70:507, 1969.
  363. Viherkoski M, Lamberg BA: The glucose-6-phosphate dehydrogenase activity (G-6-PD) of the red blood cells in hyperthyroidism and hypothyroidism. Scand J Clin Lab Invest 25:137, 1970.
  364. Irvine WJ, Wu FCW, Urbaniak SJ, Toolis F: Peripheral blood leukocytes in thyrotoxicosis (Graves' Disease) as studied by conventional light microscopy. Clin Exp Immunol 27:216, 1977.
  365. Hertz S, Lerman J: The blood picture in exophthalmic goiter and its changes resulting from iodine and operation. J Clin Invest 11:1179, 1932.
  366. Lima CS, Wittmann DE, Castro V, Tambascia MA, Lorand-Metze I, Saad ST, Costa FF ..Pancytopenia in untreated patients with graves' disease. Thyroid. 2006 Apr;16(4):403-409.
  367. Lamberg BA, Kivikangas V, Pelkonen R, Viopio P: Thrombocytopenia and decreased life-span of thrombocytes in hyperthyroidism. Ann Clin Res 3:98, 1971.
  368. Adrouny A, Sandler RM, Carmel R: Variable presentation of thrombo- cytopenia in Graves' disease. Arch Intern Med 142:1460-1464, 1982.
  369. Verberne HJ, Fliers E, Prummel MF, Stam J, Brandjes DP, Wiersinga WM. Thyrotoxicosis as a predisposing factor for cerebral venous thrombosis. Thyroid 10:607, 2000.
  370. Bergman TA, Mariash CN, Oppenheimer JH: Anterior mediastinal mass in a patient with Graves' disease. J Clin Endocrinol Metab 55:587, 1982.
  371. Okumura M, Hidaka Y, Kuroda S, Takeoka K, Tada H, Amino N. Increased serum concentration of soluble CD30 in patients with Graves’ disease and Hashimoto’s thyroiditis. J Clin Endocrinol Metab 82:1757-1760, 1997
  372. Lerman J, Means JH: The gastric secretion in exophthalmic goiter and myxedema. J Clin Invest 11:167, 1932.
  373. Berryhill WR, Williams HA: A study of the gastric secretion in hyper- thyroidism before and after operation. J Clin Invest 11:753, 1932.
  374. Siurala M, Lamberg BA: Stomach in thyrotoxicosis. Acta Med Scand 165:181, 1959.
  375. Seino Y, Matsukura S, Miyamoto Y, Goto Y, Taminato T, Imura H: Hyper- gastrinemia in hyperthyroidism. J Clin Endocrinol Metab 43:852, 1976.
  376. Wegener M, Wedmann B, Langhoff T, Schaffstein J, Adamek R. Effect of hyperthyroidism on the transit of a caloric solid-liquid meal through the stomach, the small intestine, and the colon in man. J Clin Endocrinol Metab 75:745-749, 1992.
  377. Lamberg BA, Gordin R: Liver function in thyrotoxicosis. Acta Endocrinol 15:82, 1954.
  378. Thompson Jr P, Strum D, Boehm T, Wartofsky L: Abnormalities of liver function tests in thyrotoxicosis. Military Medicine 548-551, 1978.
  379. Klion FM, Segal R, Schaffner F: The effect of altered thyroid function on the ultrastructure of the human liver. Am J Med 50:317, 1971.
  380. Greenberger NJ, Milligan FD, DeGroot LJ, Isselbacher KJ: Jaundice and thyrotoxicosis in the absence of congestive heart failure. Am J Med 36:840, 1964.
  381. Ford RV, Owens JC, Curd GW Jr, Moyer JH, Spurr CL: Kidney function in various thyroid states. J Clin Endocrinol Metab 21:548, 1961.
  382. Epstein FH, Rivera MJ: Renal concentrating ability in thyrotoxicosis. J Clin Endocrinol Metab 18:1135, 1958.
  383. Huth EJ, Maycock RL, Kerr RM: Hyperthyroidism associated with renal tubular acidosis. Am J Med 26:818, 1959.
  384. Goldsmith RE, Sturgis SH, Lerman J, Stanbury JB: The menstrual pattern in thyroid disease. J Clin Endocrinol Metab 12:846, 1952.
  385. Bray GA, Jacobs HS: Thyroid activity and other endocrine glands. In: Handbook of Physiology, Endocrinology, Washington, DC, American Physiology Society, 1974, vol III, p 413.
  386. Freedberg IM, Hamolsky MW, Freedberg AS: The thyroid gland in pregnancy. N Engl J Med 256:505, 1957.
  387. Abalovich M, Amino N, Barbour LA, Cobin RH, De Groot LJ, Glinoer D, Mandel SJ, Stagnaro-Green A. Management of thyroid dysfunction during pregnancy and postpartum: an Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2007 Aug;92(8 Suppl):S1-47
  388. Anselmo J, Cao D, Karrison T, Weiss RE, Refetoff S. Fetal loss associated with excess thyroid hormone exposure.JAMA. 2004 Aug 11;292(6):691-5.
  389. Poppe K, Glinoer D, Van Steirteghem A, Tournaye H, Devroey P, Schiettecatte J, Velkeniers B. Thyroid dysfunction and autoimmunity in infertile women. Thyroid. 2002 Nov;12(11):997-1001.
  390. Negro R, Formoso G, Mangieri T, Pezzarossa A, Dazzi D, Hassan H. Levothyroxine treatment in euthyroid pregnant women with autoimmune thyroid disease: effects on obstetrical complications. J Clin Endocrinol Metab. 2006 Jul;91(7):2587-91.
  391. Polak M, Le Gac I, Vuillard E, Guibourdenche J, Leger J, Toubert ME, Madec AM, Oury JF, Czernichow P, Luton D. Fetal and neonatal thyroid function in relation to maternal Graves' disease. Best Pract Res Clin Endocrinol Metab. 2004 Jun;18(2):289-302
  392. Becker KL, Winnacker JL, Matthews MJ, Higgins GA: Gynecomastia and hyperthyroidism. An endocrine and histological investigation. J Clin Endocrinol Metab 28:277, 1968.
  393. Chopra IJ, Tulchinsky D: Status of estrogen-androgen balance in hyper- thyroid men with Graves' disease. J Clin Endocrinol Metab 38:269, 1974.
  394. Chopra IJ, Abraham GE, Chopra U, Solomon DH, O'Dell WD: Alterations in circulating estradiol-17-beta in male patients with Graves' disease. N Engl J Med 286:124, 1972.
  395. Bercovici JP, Mauvais-Jarvis P: Hyperthyroidism and gynecomastia: Metabolic studies. J Clin Endocrinol Metab 35:671, 1972.
  396. Southren AL, Olivo J, Gordon GG, Vittek J, Brener J, Rafii F: The conversion of androgens to estrogens in hyperthyroidism. J Clin Endocrinol Metab 38:207, 1974.
  397. Kidd SG, Glass AR, Vigersky RA: The hypothalamic-pituitary- testicular axis in thyrotoxicosis. J Clin Endocrinol 48:798, 1979.
  398. Krassas, GE; Perros, P. Thyroid disease and male reproductive function. J Endocrinol Invest 26 372-380 2003
  399. Abalovich M, Levalle O, Hermes R, Scaglia H, Aranda C, Zylbersztein C, et al. 1999 Hypothalamic-pituitary-testicular axis and seminal parameters in hyperthyroid males. Thyroid 9:857262.2. In thyrotoxicosis, mean sperm density is lower, and fewer sperm have normal morphology. Motility is lower in thyrotoxic males. The abnormalities normalize when the patients become euthyroid.
  400. Sawers JSA, Kellett HA, Brown NS, Seth J, Toft AD: Prolactin response to metoclopramide in hyperthyroidism. J Clin Endocrinol Metab 55:175, 1982.
  401. Kapcala LP: Galactorrhea and thyrotoxicosis. Arch Intern Med 144:2349-2350, 1984.
  402. Peterson RE: The influence of the thyroid on adrenal cortical function. J Clin Invest 37:736, 1958.
  403. Kenny FM, Iturzaeta N, Preeyasombat C, Taylor FH, Migeon CJ: Cortisol production rate. VII. Hypothyroidism and hyperthyroidism in infants and children. J Clin Endocrinol 27:1616, 1967.
  404. Hellman L, Bradlow HL, Zumoff B, Gallagher TF: Influence of thyroid hormone on hydrocortisone production and metabolism. 21:1231, 1961.
  405. Gallagher TF, Hellman L, Finkelstein J, Yoshida K, Weitzman ED, Roffwarg HD, Fukushima D: Hyperthyroidism and cortisol secretion in man. J Clin Endocrinol Metab 34:919, 1972.
  406. Hilton JG, Black WC, Athos W, McHugh B, Westermann CD: Increased ACTH-like activity in plasma of patients with thyrotoxicosis. J Clin Endocrinol Metab 22:900, 1962.
  407. Felber JP, Reddy WJ, Selenkow HA, Thorn GW: Adrenocortical response to the 48-hour ACTH test in myxedema and hyperthyroidism. J Clin Endocrinol Metab 19:895, 1959.
  408. Hellman L, Bradlow HL, Zumoff B, Fukushima DK, Gallagher TF: Thyroid-androgen interrelations and the hypocholesteremic effect of androsterone. J Clin Endocrinol Metab 19:936, 1959.
  409. Plummer HS, Boothby WM: The cost of work in exophthalmic goiter. Am J Physiol 53:406, 1923.
  410. Briard SP, McClintock JT, Baldridge CW: Cost of work in patients with hypermetabolism due to leukemia and to exophthalmic goiter. Arch Intern Med 56:30, 1935.
  411. Acheson K, Jequier E, Burger A, Danforth Jr E: Thyroid hormones and thermogenesis: The metabolic cost of food and exercise. Metabolism 33:262-265, 1984.
  412. Kabadi UM, Eisenstein AB: Impaired pancreatic alpha-cell response in hyperthyroidism. J Clin Endocrinol Metab 51:478, 1980.
  413. Wennlund A, Arner P, Ostman J: Changes in the effects of insulin on human adipose tissue metabolism in hyperthyroidism. J Clin Endocrinol Metab 53:631, 1981.
  414. Houssay BA: Thyroid and metathyroid diabetes. Endocrinology 35:158,1944.
  415. Bratusch-Marrain PR, Komjati M, Waldhausal WK: Glucose metabolism in noninsulin-dependent diabetic patients with experimental hyperthyroidism. J Clin Endocrinol Metab 60:1063, 1985.
  416. Elrick H, Hlad CJ Jr, Arai Y: Influence of thyroid metabolism on carbohydrate metabolism and a new method for assessing response to insulin. J Clin Endocrinol Metab 21:387, 1961.
  417. Gerstein HC. Incidence of postpartum thyroid dysfunction in patients with type I diabetes mellitus. Annals Int Med 118:419-423, 1993.
  418. Kritchevsky D: Influence of thyroid hormones and related compounds on cholesterol biosynthesis and degradation: A review. Metabolism 9:984, 1960.
  419. Siperstein MD, Murray AW: Cholesterol metabolism in man. J Clin Invest 34:1149, 1955.
  420. O’Brien T, Katz K, Hodge D, Nguyen TT, Kottke BA, Hay ID. The effect of the treatment of hypothyroidism and hyperthyroidism on plasma lipids and apolipoproteins AI, AII, and E. Clin Endocrinol 46:17-20, 1997.
  421. Chait A, Bierman EL, Albers JJ: Regulatory role of triiodothyronine in the degradation of low density lipoprotein by cultured human skin fibroblasts. J Clin Endocrinol Metab 48:887, 1979.
  422. Tulloch BR, Lewis B, Fraser RT: Triglyceride metabolism in thyroid disease. Lancet 1:391, 1973.
  423. Cachefo A, Boucher P, Vidon C, Dusserre E, Diraison F, Beylot M. Hepatic lipogenesis and cholesterol synthesis in hyperthyroid patients. J Clin Endocrinol Metab 86:5353-5357, 2001.
  424. Arons DL, Schreibman PH, Downs P, Braverman LE, Arky RA: Decreased postheparin lipases in Graves' disease. N Engl J Med 286:233, 1972.
  425. Sachs BA, Danielson E, Isaacs MC, Weston RE: Effect of triiodothyronine on the serum lipids and lipoproteins of euthyroid and hyperthyroid subjects. J Clin Endocrinol Metab 18:506, 1958.
  426. Kung AW, Pang RW, Lauder I, Lam KS, Janus ED. Changes in serum lipoprotein(a) and lipids during treatment of hyperthyroidism. Clin Chem 41:226-231, 1995.
  427. Ozata M, Uckaya G, Bolu E, Corapcioglu D, Bingol N, Ozdemir IC. Plasma leptin concentrations in patients with Graves’ disease with or without ophthalmopathy. Medical Science Monitor 7:696-700, 2001.
  428. Strisower B, Elmlinger P, Gofman JW, deLalla O: Effect of L-thyroxine on serum lipoprotein and cholesterol concentrations. J Clin Endocrinol Metab 19:117, 1959.
  429. Postel S: Total free tocopherols in the serum of patients with thyroid disease. J Clin Invest 35:1345, 1956.
  430. Rich C, Bierman EL, Schwartz I: Plasma nonesterified fatty acids in hyperthyroid states. J Clin Invest 38:275, 1959.
  431. Lewallen CG, Rall JE, Berman M: Studies of iodoalbumin metabolism. II. The effects of thyroid hormone. J Clin Invest 38:88, 1959.
  432. Crispell KR, Parson W, Hollifield G: A study of the rate of protein synthesis before and during the administration of L-triiodothyronine to patients with myxedema and healthy volunteers using N-15 glycine. J Clin Invest 35:164, 1956.
  433. Sokoloff L, Kaufman S: Effects of thyroxine on amino acid incorporation into protein. Science 129:569, 1959.
  434. Sokoloff L, Kaufman S, Gelboin HV: Thyroxine stimulation of soluble ribonucleic acid bound amino acid transfer to microsomal protein. Biochim Biophys Acta 52:410, 1961.
  435. Kivirikko KI, Laitinen O, Aer J, Halme J: Metabolism of collagen in experimental hyperthyroidism and hypothyroidism in the rat. Endocrinology 80:1051, 1967.
  436. Singh SP, Snyder AK: Effect of thyrotoxicosis on gluconeogenesis from alanine in the perfused rat liver. Endocrinology 102:182, 1978.
  437. Benvenga S, Ruggeri RM, Russo A, Lapa D, Campenni A, Trimarchi F. Usefulness of L-carnitine, a naturally occurring peripheral antagonish of thyroid hormone action, in iatrogenic hyperthyroidism: a randomized, double-blind, placebo-controlled clinical trial. J Clin Endocrinol Metab 86:3579-3594, 2001.
  438. Guttler RB, Croxson MS, DeQuattro VL, Warren DW, Otis CL, Nicoloff JT: Effects of thyroid hormone on plasma adenosine 3',5'-monophosphate production in man. Metabolism 26:1155, 1977.
  439. Nilsson OR, Andersson RGG, Karlberg BE: Effects of propranolol and atenolol on plasma and urinary cyclic adenosine 3',5'-monophosphate in hyperthyroid patients. Acta Endocrinol 94:38, 1980.
  440. Peracchi M, Bamonti-Catena F, Lombardi L, Reschini E, Toschi V, Maiolo AT, Polli EE: Plasma and urine cyclic nucleotide levels in patients with hyperthyroidism and hypothyroidism. J Endocrinol Invest 6:173, 1983.
  441. Wohl MG, Levy HA, Szutka A, Maldia G: Pyridoxine deficiency in hyperthyroidism. Proc Soc Exp Biol Med 105:523, 1960.
  442. Stein JA, Griem ML: Effect of triiodothyronine on radiosensitivity. Nature 182:1681, 1958.
  443. Parker JLW, Lawson DH: Death from thyrotoxicosis. Lancet 2:894, 1973.
  444. Davis PJ, Davis FB: Hyperthyroidism in patients over the age of 60 years. Medicine 53:161, 1974.
  445. Wong GWK, Lai J, Cheng PS. Growth in childhood thyrotoxicosis. Eur J Pediatr 158:776-779, 1999.
  446. Segni M, Leonardi E, Mazzoncini B, Pucarelli I, Pasquino AM. Special features of Graves' disease in early childhood. Thyroid 9:871, 1999.

Dyslipidemia in Chronic Kidney Disease

Recieved 2/23/15

 Abstract

Chronic kidney disease (CKD) is associated with a dyslipidemia comprising high triglycerides, low HDL-cholesterol and altered lipoprotein composition. Cardiovascular diseases are the leading cause of mortality in CKD, especially in end stage renal disease patients. Thus, therapies to reduce cardiovascular risk are urgently needed in CKD. Robust clinical trial evidence has found that use of statins in pre-end stage CKD patients, as well as in renal transplant recipients, can decrease cardiovascular events; however, providers need to be aware of dose restrictions for statin therapy in CKD subjects. Furthermore, statin therapy does not reduce cardiovascular events in dialysis patients, nor does statin therapy confer any protection against progression of renal disease. Niacin and fibrates are effective in lipid lowering in CKD and appear to have some cardiovascular benefit but further study is needed to clearly define their role. This article reviews the epidemiology of CKD, association of CKD with cardiovascular events, and the effects of CKD on lipid levels and metabolism. The article discusses clinical trial evidence for and against statin and non-statin lipid lowering therapy in CKD patients.

 CKD epidemiology

Chronic kidney disease (CKD) is defined as renal impairment greater than 3 months duration that results in an estimated glomerular filtration rate (eGFR) < 60ml/min/1.73m2. CKD is classified into 5 stages based on the eGFR (Table 1). CKD is a world-wide health problem with rising incidence and prevalence. CKD, especially in the early stages is often asymptomatic; thus, the actual prevalence may be even higher than estimated. End stage renal disease (ESRD) is defined as needing dialysis or transplant, and the prevalence and incidence of ESRD have doubled over the past 10 years(1). The annual mortality rate of dialysis patients is greater than 20%. The burden of co-morbidities and the cost of caring for CKD patients is high, and thus a major focus is increased screening and early detection of CKD when interventions to delay or prevent progression to ESRD may be effective. There are multiple causes of CKD with the most common causes in westernized nations being hypertension and diabetes; however, a wide range of etiologies including infectious, auto-immune, genetic, obstructive, and ischemic injury are all prevalent. There are ethnic differences in susceptibility with increased prevalence in Mexican-Americans and non-Hispanic blacks compared to Caucasians(2).

 

While the burden of CKD itself is significant, the leading causes of morbidity and mortality in CKD are cardiovascular diseases (CVD), primarily atherosclerotic coronary artery disease. Risk factors for CVD in CKD include the traditional risk factors – hypertension, sex, age, smoking, and family history and CKD patients appear to benefit similar to non-CKD patients from therapies targeting these risk factors. Regardless of the cause of CKD, patients with CKD are at increased risk for CVD, which has led to the National Kidney Foundation classifying all patients with CKD as “highest risk” for CVD regardless of their levels of traditional CVD risk factors. The focus of this chapter is on the dyslipidemia of CKD and the risk of CVD in CKD.

 Nephrotic syndrome:
Nephrotic syndrome differs from other types of CKD in its presentation and risks. Nephrotic syndrome is comprised of significant proteinuria (typically > 3g/24h), hypoalbuminemia, peripheral (+/- central) edema, and significant hyperlipidemia and lipiduria may also be seen. It is frequently seen in children, and the etiology includes minimal change disease (up to 85%), focal segmental glomerulosclerosis (up to 15%) and secondary causes (rare) including systemic lupus erythrematosis, Henoch Schonlein Purpura, or membrano-proliferative glomerulopathy. In adults, the etiology is more likely to involve a systemic disease such as diabetes, amyloidosis, or lupus. Nephrotic syndrome may be transient or persistent. Most (approximately 80% of children) cases of nephrotic syndrome are successfully treated with glucocorticoids with resolution of all features including hyperlipidemia; however, steroid-resistant nephrotic syndrome patients often have persistent dyslipidemia, which may place them at increased risk for CVD. For example a small study found increased CVD markers including pulse wave velocity, carotid artery intima-media thickness and left ventricular mass in patients with steroid-resistant nephrotic syndrome compared to controls(3), implying increased risk for CVD events. Treatment of nephrotic syndrome dyslipidemia includes therapies specifically targeting the renal disease (primarily glucocorticoids, but also renin-angiotensin system antagonists which can help decrease proteinuria) and lipid lowering agents.

 Table 1: Stages of CKD

CKD stage GFR (ml/min/1.73 m2)
CKD 1 ≥ 90 (with renal damage or injury)
CKD 2 (mild) 60-89
CKD 3 (moderate) 30-59
CKD 4 (severe) 15-29
CKD 5 (end stage) <15, dialysis, or transplant

CVD IN CKD

CVD accounts for 40-50% of all deaths in ESRD patients, with CVD mortality rates approximately 15 times that seen in the general population(4). However, CVD is highly prevalent in patients who progress to ESRD implying that earlier stages of CKD increase the development of CVD. A number of factors have been proposed as risk factors for CVD in CKD including proteinuria, inflammation, anemia, malnutrition, oxidative stress and uremic toxins(5). Ongoing research is investigating whether these (and other) markers may be therapeutic targets. Interestingly, proteinuria correlates with blood pressure, total cholesterol, triglycerides, and inversely correlates with HDL-cholesterol(6). Thus, it remains unclear if proteinuria itself is a risk factor (e.g. a cause of CVD) or a biomarker. Meta-analyses of general population and high risk population cohorts found that both lower eGFR (<60 ml/min/1.73 m2) and higher albuminuria (>10 mg/g creatinine) are predictors of total mortality and CVD mortality;  furthermore, eGFR and albuminuria are independent of each other and of traditional CVD risk factors (7, 8). Estimated GFR > 60 ml/min/1.73 m2 is not a risk factor for CVD or total mortality.

Dyslipidemia in CKD

Effect of CKD on lipid levels:
CKD is associated with a dyslipidemia comprised of elevated triglycerides and low HDL-cholesterol. Levels of LDL-cholesterol (and thus, total cholesterol) are generally not elevated; however, proteinuria correlates with cholesterol and triglycerides. CKD leads to a down regulation of lipoprotein lipase and the LDL-receptor, and increased triglycerides in CKD are due to delayed catabolism of triglyceride rich lipoproteins, with no differences in production rate(9). CKD is associated with lower levels of apoA-I (due to decreased hepatic expression(10)) and higher apoB/apoA-I. Decreased lecithin-cholesterol acyltransferase (LCAT) activity and increased cholesteryl ester transfer protein (CETP) activity contribute to decreased HDL-cholesterol levels. Beyond decreased HDL cholesterol levels, the HDL in CKD is less effective in its anti-oxidative and anti-inflammatory functions [for review see (11)].

 

As CKD progresses the dyslipidemia often worsens. In an evaluation of 2001-2010 National Health and Nutrition Examination Survey (NHANES), the prevalence of dyslipidemia increased from 45.5% in CKD stage 1 to 67.8% in CKD stage 4; similarly, the use of lipid lowering agents increased from 18.1% in CKD stage 1 to 44.7% in CKD stage 4(12).  Of more than 1000 hemodialysis patients studied only 20% had “normal” lipid levels (defined as LDL<130 mg/dl, HDL > 40 and triglycerides < 150); of 317 peritoneal dialysis patients only 15% had “normal” lipid levels(13). A larger study evaluating dyslipidemia in > 21,000 incident dialysis patients found 82% prevalence of dyslipidemia and suggested a threshold of non-HDL cholesterol > 100 mg/dl (2.6mmol/L) to identify dyslipidemia in CKD stage 5 subjects(14). Peritoneal dialysis is associated with higher cholesterol levels than hemodialysis, although the reasons aren’t fully understood. In subjects who switched from peritoneal dialysis to hemodialysis there was a drop in cholesterol levels of almost 20% following transition (15). The National Kidney Foundation recommends routine screening of all adults and adolescents with CKD using a standard fasting lipid profile (total cholesterol, LDL-cholesterol, HDL-cholesterol and triglycerides), and follows the classification of the National Cholesterol Education Panel for levels (desirable, borderline or high). Although some studies have found associations between Lp(a) and dialysis patients, this is not well defined and there is no current indication for routine screening of Lp(a).

Effect of CKD on lipoprotein composition:

Beyond simply measuring lipid levels, emerging evidence implies that lipoprotein particle size and composition is altered in CKD, with increased small dense LDL and decreased larger LDL particles in CKD subjects compared to controls(16). Small dense LDL is thought to be more atherogenic than larger LDL particles. An emerging theory is that beyond lipid levels or lipoprotein size, lipoprotein particle “cargo” can affect atherosclerosis development and progression. Lipoprotein particles transport numerous bioactive lipids, microRNAs, other small RNAs, proteins, hormones, etc. For example, a recent study compared LDL particle composition between subjects with stage 4/5 CKD and non-CKD controls, and found similar total lipid and cholesterol content, but altered content of various lipid subclasses, for example decreased phosphatidylcholines, sulfatides and ceramides and increased N-acyltaurines(17). Many of these lipid species are known to have either pro- or anti-atherogenic properties and thus could directly affect atherogenesis.

Effect of renal transplantation on lipid levels
:
Dyslipidemia is frequently seen in renal transplant recipients, including increased total cholesterol, LDL-cholesterol, and triglycerides, and decreased HDL-cholesterol. The dyslipidemia may have existed pre-transplant or be related to transplantation associated factors. Cyclosporine increases LDL-cholesterol via both increased production and decreased clearance. Corticosteroids increase both cholesterol and triglyceride levels in a dose-dependent manner. The adverse effects of cyclosporine and corticosteroids on lipid levels appear to be additive(18). Tacrolimus and azathioprine appear to have less induction of dyslipidemia than cyclosporine(19). Sirolimus increases both cholesterol and triglycerides, in part due to decreased LDL-clearance(20).

 Effect of nephrotic syndrome on lipid levels:
The dyslipidemia in nephrotic syndrome can be striking with significant elevations of cholesterol, LDL-cholesterol, triglycerides and lipoprotein(a); HDL cholesterol is often low, especially HDL2. The cause of elevated lipid levels is multi-factorial, including reduction in oncotic pressure which stimulates apoB synthesis (although the exact mechanism by which this occurs is not known), decreased metabolism of lipoproteins, and decreased clearance. Patients with nephrotic syndrome have decreased LDL-receptor activity and increased acyl-CoA cholesterol acytransferase (ACAT) and HMG-CoA reductase activity leading to increased LDL-cholesterol levels(21, 22). Low HDL-cholesterol is thought to be due at least in part to LCAT deficiency secondary to accelerated renal loss of LCAT(23). Triglycerides are elevated due to impaired clearance of chylomicrons and triglyceride-rich lipoproteins, as well as increased triglyceride production(24).

Evidence for/against lipid lowering therapy in CKD for CVD outcomes

Given the high prevalence of CVD in CKD, and the robust clinical evidence in non-CKD subjects that lipid lowering reduces CVD outcomes, there is great interest in using lipid lowering therapy in CKD subjects. Statins are the most commonly used lipid-lowering medications and thus far have been shown to reduce CVD events and/or mortality in virtually every population studied. However, CKD patients seem to be a unique population in that at present there is no evidence of benefit for CVD outcomes in dialysis patients with statin therapy. As discussed below it appears that statins can reduce CVD events in pre-end stage CKD subjects, and in post-renal transplant subjects, but not in dialysis patients (Table 2).

Use of statins in pre-ESRD CKD patients:
Although many of the initial statin CVD studies did not include many CKD patients, evidence from sub-group analyses of large statin studies suggested that CKD subjects had similar benefits to non-CKD individuals. For example, the Heart Protection Study (HPS) which assessed >20,000 subjects at high risk of CVD included a subgroup of 1329 subjects with impaired kidney function. In this subgroup those that received simvastatin had a 28% proportional risk reduction and an 11% absolute risk reduction of a major cardiovascular event compared to those randomized to placebo; similar to the effect on the overall cohort(25). Several other studies or meta-analyses similarly predicted that CKD subjects would have reduction in CVD with statin therapy. For example, a meta-analysis of 38 studies with >37,000 participants with CKD but not yet on dialysis found a consistent reduction in major cardiovascular events, all-cause mortality, cardiovascular death and myocardial infarction in statin users compared to placebo groups. There was no clear effect of statin on stroke, nor was there any effect of statin use on progression of the renal disease(26). Thus, for CKD patients with pre-end stage renal disease statins effectively lower total cholesterol and LDL-cholesterol levels and decrease CVD risk. The different statins have different degrees of renal involvement in their metabolism, and providers should be aware of dose restrictions in CKD (Table 3).

Unclear whether to use statins in subjects with nephrotic syndrome:
Several small clinical studies have investigated the use of lipid lowering therapies in nephrotic syndrome, but data is only available for statins and fibrates, and no CVD outcomes data is available. Several small studies using statins have found efficacy in lowering LDL-cholesterol, and that statins were safe and well tolerated(27, 28). Thus, the use of statins in nephrotic syndrome appears to be safe and efficacious in terms of lipid lowering; however, it is not clear if there is any corresponding benefit on either CVD or renal outcomes.

 No benefit to statins in subjects with only microalbuminuria:
The Prevention of Renal and Vascular Endstage Disease Intervention Trial (PREVEND IT) randomized 864 subjects with persistent microalbuminuria (urinary albumin of 15-300mg/24h x 2 samples) to fosinopril (an angiotensin converting enzyme inhibitor) or placebo and to pravastatin 20 mg or placebo. Inclusion criteria for the study included blood pressure <160/100 mm Hg and no use of antihypertensive medications and total cholesterol < 300 mg/dl (8 mmol/L) or < 192 mg/dl (5 mmol/L) if patient had known CVD and no use of lipid lowering medications. Although diabetes was not an exclusion criteria, <3% of the subjects had diabetes(29). The use of statin did not affect either urinary albumin excretion or cardiovascular events; however, the use of fosinopril significantly decreased albuminuria and had a trend to reduction in cardiovascular events. Thus, in the absence of other indications for statin therapy, there appears to be no benefit in subjects that solely have microalbuminuria. However, a subsequent analysis found that the subjects with isolated microalbuminuria had an increased risk for CVD events and mortality compared to those without risk factors(30); thus isolated microalbuminuria appears to indicate high risk and further study is needed to determine effective therapies to reduce risk.

No benefit to statins in dialysis patients:

Studies specifically examining the role of statins in ESRD subjects have not found a benefit. The Deutsche Diabetes Dialyse Studie (4D) randomized 1255 type 2 diabetic subjects on maintenance hemodialysis to either 20 mg atorvastatin or placebo daily. The cholesterol and LDL-cholesterol reduction was similar to that seen in non-dialysis patients; however, unlike non-CKD subjects there was no significant reduction in cardiovascular death, nonfatal myocardial  infarction or stroke with atorvastatin compared to placebo(31). Similarly, A Study to Evaluate the Use of Rosuvastatin in Subjects on Regular Hemodialysis (AURORA) randomized 2776 subjects on maintenance hemodialysis to rosuvastatin 10 mg or placebo. Again, the LDL-cholesterol lowering in dialysis patients was similar to that seen in other studies in non-dialysis patients, but there was no significant effect on the primary endpoint of cardiovascular death, nonfatal myocardial infarction or stroke(32). The Study of Heart and Renal Protection (SHARP) randomized 9270 CKD patients (3023 on dialysis) to simvastatin plus ezetimibe versus placebo. Unlike 4D and AURORA, the SHARP study did report a significant reduction in major atherosclerotic events in the simvastatin plus ezetimibe group, including the dialysis subgroup(33). However, a meta-analysis of 25 studies involving 8289 dialysis patients found no benefit of statin therapy on major cardiovascular events, cardiovascular mortality, all-cause mortality or myocardial infarction, despite efficacious lipid lowering(34). Nevertheless, a post-hoc analysis of the 4D study did demonstrate a benefit of statin therapy in the subgroup that had LDL cholesterol > 145 mg/dl (3.76mmol/l) (35). Although the use of statins in dialysis patients does not clearly cause harm, at present there is no indication for use in dialysis patients, with the exception of a possible benefit in those with significant elevation in LDL-cholesterol.

Why is statin therapy ineffective in dialysis subjects?
Given the robust data demonstrating statin efficacy in CVD risk reduction in virtually all other populations studied, the lack of efficacy in ESRD subjects in perplexing. However, it may be due to different mechanisms of disease progression in ESRD populations compared to other populations. In ESRD subjects there is increased inflammation and oxidative stress as well as increased non-lipid-associated pro-atherogenic factors, which may be the major cause of atherosclerosis development or progression in CKD subjects [for review see (36)]. Therefore, the relative impact of dyslipidemia on CVD development and progression in ESRD subjects may be less than in other CKD and non-CKD subjects, and thus the potential benefit of lipid lowering therapy is reduced. In ESRD subjects with significant hyperlipidemia (such as genetic hyperlipidemias) there may still be a role for statins, or other lipid lowering therapies. Furthermore, while no benefit has been found for statins in dialysis subjects, there is no evidence of increased harm, and thus consideration of lipid lowering medications in particular individuals with ESRD is warranted.

Use of statins in renal transplant recipients:
The Assessment of Lescol in Renal Transplant (ALERT) study randomized 2102 renal transplant recipients to fluvastatin or placebo. There was a non-significant 17% reduction in the combined primary endpoint (cardiac mortality, nonfatal myocardial infarction or coronary intervention procedures) but a significant reduction in cardiac death or myocardial infarction(37, 38). Furthermore, a post hoc analysis suggested that earlier initiation of statins post-transplant was associated with greater benefit(39). However, as with pre-end stage CKD patients there did not appear to be any benefit from statin therapy on progression of renal disease or graft loss in statin treated transplant recipients. (40) Thus, following renal transplant patients should be considered for statin therapy for CVD risk reduction, but not for graft preservation. Several of the statins have drug interactions, particularly with cyclosporine, thus providers must be aware of dose and drug restrictions (Table 3).

Table 2: Use of statins in various CKD subgroups:

Patient population Statin indicated? Yes/no
Microalbuminuria* No
CKD 1-4 Yes
Nephrotic syndrome Unclear
Dialysis patients No
Renal transplant recipients Yes

* in the absence of any other indication

Evidence for/against lipid lowering therapy in CKD for renal outcomes

Given the evidence that renal lipid deposition is associated with progression of renal disease itself, there has been an ongoing interest in whether targeting dyslipidemia in CKD can help delay the progression of the renal disease. The dyslipidemia in CKD is associated not only with increased CVD but also with adverse renal prognosis(41, 42). Biopsy studies have found that the amount of renal apoB/apoE is correlated with increased progression of the renal disease itself(43). Animal studies have supported this concept. A meta-analysis of several small, older studies suggested that the rate of decline in GFR was decreased in subjects receiving a lipid-lowering agent (the included studies mainly used statins but the meta-analysis also included a study using gemfibrozil and another using probucol)(44). However, the relationship between lipid levels and renal disease is unclear, as prospective cohort studies have not found any relationship of lipid levels to progression of kidney disease(45). Furthermore, the SHARP study, which included subjects with earlier stages of CKD (stages 3-5 were included) found no benefit of lipid lowering therapy on the progression of renal disease. A meta-analysis of statins in pre-end stage CKD patients found no overall effect of statins on renal disease progression(26) and the ALERT study found no benefit of statin use on renal graft or renal disease parameters(40). Thus, there does not appear to be any use for statins to improve renal function or CKD itself.

Safety of statins in CKD

Statin safety– renal outcomes:
A recent observational study using administrative databases containing information on > 2 million patients suggested that the use of high potency statins was associated with acute kidney injury, especially within the first 120 days of statin use(46). However, a subsequent analysis of 24 placebo controlled statin studies and 2 high versus low-dose statin studies found no evidence of renal injury from statin use(47). These discrepant results can be explained by the quality of the data: in randomized controlled trials, albeit not designed or powered to look at renal injury, data quality tends to be higher than that in administrative data sets, which often contain bias for selection, ascertainment and classification. Furthermore, statins appear to have a nephron-protective role in the prevention of contrast induced acute kidney injury. A meta-analysis of 15 trials examining the effect of statin pre-treatment before coronary angiography found a significant reduction in acute kidney injury in those treated with high dose statin compared to controls treated with either placebo or low dose statin(48). One study specifically examined the use of statins in subjects with diabetes and existing CKD undergoing angiography, and found a benefit to statin pre-treatment in reducing the risk of contrast induced acute renal injury(49). As discussed above, use of statins in pre-end stage CKD or post-renal transplant patients demonstrates neither benefit nor harm on renal outcomes. Thus, based on available evidence there does not seem to be any renal harm from statin use, and the presence of CKD should not be a contra-indication to statin use, although some statins require dose restrictions in CKD (Table 3).

Statin safety – diabetes outcomes:
As a class, emerging evidence demonstrates that statins increase new diagnoses of diabetes(50). As diabetes can lead to or exacerbate renal injury, this is another potential harm of statins. A subsequent meta-analysis of 5 statin trials with >32,000 patients without diabetes at baseline found that high dose statin was associated with increased risk for new diabetes compared to low or moderate dose statin therapy(51). However, the number needed to harm (induce diabetes) is 498 whereas the number needed to treat (prevent cardiovascular events) is 155 for intensive statin therapy; implying that despite the increased risk of new onset diabetes, statin therapy’s benefits outweigh the risks.

Which statins to use in CKD?

The various statins have different degrees of renal clearance; thus, with CKD patients it is important to be aware of the metabolism of the agent of interest and understand if/when dose adjustments are needed. Most statins are primarily metabolized through hepatic pathways, and dose adjustment in early CKD is typically not needed (eGFR> 30 ml/min). However, with more advanced CKD, eGFR< 30 ml/min (or ESRD, although statins are not indicated in this population) most agents have maximum dose restrictions (Table 3).

 Table 3: Statin dosing in CKD

Statin Usual dose range (mg/d) Clearance route Dose range for CKD stages1-3 Dose range for CKD stages4-5 Use with cyclosporine
Atorvastatin 10-80 Liver 10-80 10-80 Avoid use with cyclosporine
Fluvastatin 20-80 Liver 20-80 20-40 Max dose 20 mg/d with cyclosporine
Lovastatin 10-80 Liver 10-80 10-20 Avoid use with cyclosporine
Pitavastatin 1-4 Liver/Kidney 1-2 1-2 Avoid use with cyclosporine
Pravastatin 10-80 Liver/Kidney 10-80 10-20 Max dose 20 mg/d when used with cyclosporine
Rosuvastatin 10-40 Liver/Kidney 5-40 5-10 Max dose 5 mg/d with cyclosporine
Simvastatin 5-40 Liver 5-40 5-40 Avoid use with cyclosporine

Beyond statins:

There has been relatively little research into the use of non-statin lipid lowering agents in CKD. There is an emerging interest in niacin in CKD patients for its phosphorus-lowering effects, and niacin has similar lipid-altering efficacy in CKD as opposed to non-CKD subjects. Fibrates are metabolized via the kidney and thus generally contraindicated in CKD. The following sections summarize the available data on the use of other lipid lowering agents in CKD (Table 4).

Niacin:
As niacin is not cleared via the kidney it is theoretically safe in CKD; however, its use is limited due to side effects (predominantly flushing) and a lack of data. Several short term studies have evaluated niacin in CKD patients and it is efficacious in lipid lowering. There is an emerging interest in use of niacin or its analog niacinimide in CKD and ESRD patients for their effects to decrease phosphate levels. A meta-analysis of randomized controlled trials of niacin and niacinamide in dialysis patients found that niacin reduced serum phosphorus but did not change serum calcium levels; furthermore niacin increased HDL levels but had no significant effect on LDL-cholesterol, triglycerides or total cholesterol levels; no CVD outcomes data were provided(52). Animal studies have suggested a beneficial effect of niacin on renal outcomes(53), and clinical literature is suggestive that this may occur in humans. A database study of niacin in veterans reported that niacin may slow progression of chronic kidney disease (reported at the 2014 Kidney Week meeting(54)), but this data is not yet published and further study is needed. Kang et al treated patients with CKD stages 2-4 with niacin 500mg/d x 6 months; niacin led to increased HDL-cholesterol and decreased triglyceride levels, and improved GFR compared to baseline levels(55). Laropiprant has been developed as an inhibitor of prostaglandin-medicated niacin-induced flushing. In a sub-study examining the use of niacin with laropiprant in dyslipidemic subjects with impaired renal function, the use of niacin resulted in a mean decrease in serum phosphorus of 11% with similar effects between those with eGFR above or below 60 ml/min/1.73 m2(56); the parent study reported significant reduction in lipid parameters including a decrease in LDL-cholesterol of 18%, decrease in triglycerides of 25% and an increase in HDL of 20%(57). Thus, there may be an indication for use of niacin in CKD subjects beyond lipid lowering considerations. However, cardiovascular outcome studies evaluating the combination of statin plus niacin have not found any additional benefit compared to statin alone(58, 59); thus, at this time further research is needed in CKD subjects to determine if niacin may be more beneficial than statins, or if the addition of niacin to statin may confer non-CVD benefit, for example, from phosphorus lowering.

Fibrates:
Fibric acid derivatives which are used primarily to raise HDL-cholesterol and lower triglycerides; thus, they target two major components of CKD associated dyslipidemia. However, fibrates are known to decrease renal blood flow and glomerular filtration and they are cleared renally(60); therefore, there is significant concern re their use in CKD. Furthermore, the fibric acid derivatives raise serum creatinine levels and may thus trigger medical investigations into renal disease progression. Thus, there is concern regarding their use in CKD. However, there is a potential for fibric acid derivatives to improve both CVD and CKD outcomes. The acute changes in serum creatinine do not necessarily indicate adverse renal effects. A meta-analysis(61)  examined the use of fibrates in CKD subjects and reported beneficial effects to reduce total cholesterol and triglyceride levels and raise HDL-cholesterol levels with no effect on LDL-cholesterol levels. In addition, 3 trials reporting on > 14,000 patients reported that fibrates reduced risk of albuminuria progression in diabetic subjects, with 2 trials (>2,000 patients) reporting albuminuria regression (62-64). This was associated with a reduction in major cardiovascular events, CVD death, stroke and all-cause mortality in subjects with moderate renal dysfunction, but not in those with eGFR > 60 ml/min/1.73m2.  Thus, despite the elevations in serum creatinine seen with fibrates, there is the potential for both cardiac and renal benefit, and further studies specifically designed to evaluate these outcomes in CKD subjects are urgently needed. At this point, providers are encouraged to consider fibrate therapy for appropriate subjects, especially if statins are not tolerated or are contra-indicated.

 

Ezetimibe:
Ezetimibe is presently the only member of the class of cholesterol absorption inhibitors. As monotherapy it can lower LDL approximately 15%; however, the majority of research has examined ezetimibe in combination with a statin (primarily simvastatin) where the addition of ezetimibe can induce a further 25% lowering of LDL cholesterol. Ezetimibe is metabolized through intestinal and hepatic metabolism, and does not require any dose adjustment in CKD or ESRD, making it potentially attractive therapy in CKD. Recently, the results of the Improved Reduction of Outcomes: Vytorin Efficacy International Trial (IMPROVE IT) study were presented (American Heart Association Scientific Session November 2014; however, not published as of the time of writing this section) demonstrating that the combination of statin + ezetimibe led to further LDL lowering and improved CVD outcomes compared to statin alone in high risk patients(65).  The Study of Heart and Renal Protection (SHARP) compared CVD and renal effects in CKD patients treated with statin + ezetimibe versus placebo. There was a reduction in CVD events(33); however, there was no effect on renal disease progression(66). Note, neither of these studies included an ezetimibe only arm; thus, the effects of ezetimibe monotherapy on outcomes are unknown, although it can be expected to reduce CVD events in proportion to its degree of LDL-cholesterol lowering.   A small study evaluating ezetimibe monotherapy in CKD patients found it safe and effective(67). Thus, the use of ezetimibe with or without statin is likely to benefit pre-end stage CKD patients in terms of CVD outcomes (given that the impact of ezetimibe is on lowering LDL-cholesterol we can anticipate lack of CVD benefit in ESRD subjects based on the statin studies and SHARP).

Fish oil:
Omega-3 polyunsaturated fatty acids can lower triglyceride levels, making them a potential therapy in CKD. The role of fish oil/ omega-3 supplements in the general population for prevention of CVD events remains unclear, with some studies suggesting benefit but others finding no CVD protection. A recent meta-analysis found no evidence for CVD protection(68).  In CKD patients there is little data and it is conflicting. A small randomized study evaluated omega-3 fish oil supplements, coenzyme Q10, or both in subjects with CKD stage 3 for 8 weeks. The group that received the omega-3 supplements had decreased heart rate and blood pressure and triglycerides, but there was no effect on renal function (eGFR, or albuminuria)(69). Conversely, a study evaluating dietary omega-3 intake found that higher consumption was associated with reduced likelihood of CKD(70). Fish oil supplementation has not been found to have any clear benefit on hemodialysis arteriovenous graft function(71) or on cardiovascular events or mortality in hemodialysis patients(72). Thus, there is no clear benefit to the use of fish oil supplements in CKD, but further research is needed.

Bile acid resins:
The bile acid resins tend to be used less commonly than other classes of lipid lowering agents overall, and their use in CKD is limited by a lack of data. Bile acid resins as a class can lower LDL-cholesterol by 10-20% so they are less effective than statins; furthermore, they can raise triglyceride levels and their use is contra-indicated with elevated triglyceride levels, for example > 400-500 mg/dl (>4.5 – 5.6 mmol/L). Thus, overall bile acid resins are rarely used in CKD patients. However, their metabolism is intestinal and thus there are no required modifications for their use in mild-moderate CKD. Although there are no theoretical concerns regarding their use in ESRD there is no data to address safety or efficacy.

New therapies:
There are a number of new and pending therapies for the treatment of dyslipidemia, including mipomersen (an anti-sense oligonucleotide against apoB-100), lomitapide (a microsomal triglyceride transfer protein inhibitor), evolucumab and alirocumab (monoclonal antibodies against proprotein convertase subtilisin/kexin type 9 [PCSK9]) and evacetrapib and anacetrapib (cholesterol ester transfer protein [CETP] inhibitors). However, to date none of these agents have been studied in CKD; thus, there is no data available to assess their potential use in this population. Mipomersen and lomitapide are both FDA approved for use in homozygous familial hypercholesterolemia; none of the others are yet on the market.

Table 4- Non-statin treatments

Agent Usual dose range (mg/d) Clearance route Dose range for CKD stages1-3 Dose range for CKD stages4-5 Use with cyclosporine
Niaspan 500-2000 Hepatic/renal No data No data No data
Gemfibrozil 1200 Renal Avoid if creatinine > 2.0 mg/dl Avoid if creatinine > 2.0 mg/dl Cautious use
Fenofibrate 40-200 renal 40-60 avoid Cautious use
Ezetimibe 10 Intestinal/hepatic 10 10 Cautious use
Colsevelam 3750 (6 x 625 mg tablets daily) Intestinal No change unknown May reduce levels of cyclosporine
Fish oil 4000   No change Caution No data

SUMMARY:
CVD is the leading cause of mortality in CKD, and as with the non-CKD population dyslipidemia is a significant contributor. The dyslipidemia of CKD comprises primarily high triglyceride levels and low HDL-cholesterol levels; however, emerging data suggests that the composition of the lipoprotein particles is altered by CKD, and that altered composition and/or lipoprotein cargo may be a cause of the increased CVD in CKD. The use of statins has been shown to be safe and efficacious in lipid lowering in CKD, and of benefit in reducing CVD events in individuals with pre-end stage CKD, or post renal transplant, but not in dialysis patients. The various available agents have different clearance routes, and some statins need dose adjustment in CKD. In patients that cannot tolerate or who have contra-indications to statin therapy, there may be some benefit from use of fibrates or niacin, but further studies are needed to better investigate their use.

References

  1. National Kidney, F. 2012. KDOQI Clinical Practice Guideline for Diabetes and CKD: 2012 Update. Am J Kidney Dis 60: 850-886.
  2. Collins, A. J., R. N. Foley, B. Chavers, D. Gilbertson, C. Herzog, K. Johansen, B. Kasiske, N. Kutner, J. Liu, W. St Peter, H. Guo, S. Gustafson, B. Heubner, K. Lamb, S. Li, Y. Peng, Y. Qiu, T. Roberts, M. Skeans, J. Snyder, C. Solid, B. Thompson, C. Wang, E. Weinhandl, D. Zaun, C. Arko, S. C. Chen, F. Daniels, J. Ebben, E. Frazier, C. Hanzlik, R. Johnson, D. Sheets, X. Wang, B. Forrest, E. Constantini, S. Everson, P. Eggers, and L. Agodoa. 2012. 'United States Renal Data System 2011 Annual Data Report: Atlas of chronic kidney disease & end-stage renal disease in the United States. Am J Kidney Dis 59: A7, e1-420.
  3. Candan, C., N. Canpolat, S. Gokalp, N. Yildiz, P. Turhan, M. Tasdemir, L. Sever, and S. Caliskan. 2014. Subclinical cardiovascular disease and its association with risk factors in children with steroid-resistant nephrotic syndrome. Pediatric nephrology 29: 95-102.
  4. Foley, R. N., P. S. Parfrey, and M. J. Sarnak. 1998. Clinical epidemiology of cardiovascular disease in chronic renal disease. Am J Kidney Dis 32: S112-119.
  5. Parfrey, P. S., and R. N. Foley. 1999. The clinical epidemiology of cardiac disease in chronic renal failure. J Am Soc Nephrol 10: 1606-1615.
  6. Sarnak, M. J., B. E. Coronado, T. Greene, S. R. Wang, J. W. Kusek, G. J. Beck, and A. S. Levey. 2002. Cardiovascular disease risk factors in chronic renal insufficiency. Clin Nephrol 57: 327-335.
  7. Chronic Kidney Disease Prognosis, C., K. Matsushita, M. van der Velde, B. C. Astor, M. Woodward, A. S. Levey, P. E. de Jong, J. Coresh, and R. T. Gansevoort. 2010. Association of estimated glomerular filtration rate and albuminuria with all-cause and cardiovascular mortality in general population cohorts: a collaborative meta-analysis. Lancet 375: 2073-2081.
  8. van der Velde, M., K. Matsushita, J. Coresh, B. C. Astor, M. Woodward, A. Levey, P. de Jong, R. T. Gansevoort, C. Chronic Kidney Disease Prognosis, M. van der Velde, K. Matsushita, J. Coresh, B. C. Astor, M. Woodward, A. S. Levey, P. E. de Jong, R. T. Gansevoort, A. Levey, M. El-Nahas, K. U. Eckardt, B. L. Kasiske, T. Ninomiya, J. Chalmers, S. Macmahon, M. Tonelli, B. Hemmelgarn, F. Sacks, G. Curhan, A. J. Collins, S. Li, S. C. Chen, K. P. Hawaii Cohort, B. J. Lee, A. Ishani, J. Neaton, K. Svendsen, J. F. Mann, S. Yusuf, K. K. Teo, P. Gao, R. G. Nelson, W. C. Knowler, H. J. Bilo, H. Joosten, N. Kleefstra, K. H. Groenier, P. Auguste, K. Veldhuis, Y. Wang, L. Camarata, B. Thomas, and T. Manley. 2011. Lower estimated glomerular filtration rate and higher albuminuria are associated with all-cause and cardiovascular mortality. A collaborative meta-analysis of high-risk population cohorts. Kidney Int 79: 1341-1352.
  9. Chan, D. T., G. K. Dogra, A. B. Irish, E. M. Ooi, P. H. Barrett, D. C. Chan, and G. F. Watts. 2009. Chronic kidney disease delays VLDL-apoB-100 particle catabolism: potential role of apolipoprotein C-III. J Lipid Res 50: 2524-2531.
  10. Vaziri, N. D., G. Deng, and K. Liang. 1999. Hepatic HDL receptor, SR-B1 and Apo A-I expression in chronic renal failure. Nephrol Dial Transplant 14: 1462-1466.
  11. Schuchardt, M., M. Tolle, and M. van der Giet. 2015. High-density lipoprotein: structural and functional changes under uremic conditions and the therapeutic consequences. Handbook of experimental pharmacology 224: 423-453.
  12. Kuznik, A., J. Mardekian, and L. Tarasenko. 2013. Evaluation of cardiovascular disease burden and therapeutic goal attainment in US adults with chronic kidney disease: an analysis of national health and nutritional examination survey data, 2001-2010. BMC nephrology 14: 132.
  13. Longenecker, J. C., J. Coresh, N. R. Powe, A. S. Levey, N. E. Fink, A. Martin, and M. J. Klag. 2002. Traditional cardiovascular disease risk factors in dialysis patients compared with the general population: the CHOICE Study. J Am Soc Nephrol 13: 1918-1927.
  14. Pennell, P., B. Leclercq, M. I. Delahunty, and B. A. Walters. 2006. The utility of non-HDL in managing dyslipidemia of stage 5 chronic kidney disease. Clin Nephrol 66: 336-347.
  15. Rao, R., D. Ansell, J. A. Gilg, S. J. Davies, E. J. Lamb, and C. R. Tomson. 2009. Effect of change in renal replacement therapy modality on laboratory variables: a cohort study from the UK Renal Registry. Nephrol Dial Transplant 24: 2877-2882.
  16. Chu, M., A. Y. Wang, I. H. Chan, S. H. Chui, and C. W. Lam. 2012. Serum small-dense LDL abnormalities in chronic renal disease patients. British journal of biomedical science 69: 99-102.
  17. Reis, A., A. Rudnitskaya, P. Chariyavilaskul, N. Dhaun, V. Melville, J. Goddard, D. J. Webb, A. R. Pitt, and C. M. Spickett. 2014. Top-down lipidomics of low density lipoprotein reveal altered lipid profiles in advanced chronic kidney disease. J Lipid Res.
  18. Hricik, D. E., J. T. Mayes, and J. A. Schulak. 1991. Independent effects of cyclosporine and prednisone on posttransplant hypercholesterolemia. Am J Kidney Dis 18: 353-358.
  19. Marcen, R., J. Chahin, A. Alarcon, and J. Bravo. 2006. Conversion from cyclosporine microemulsion to tacrolimus in stable kidney transplant patients with hypercholesterolemia is related to an improvement in cardiovascular risk profile: a prospective study. Transplantation proceedings 38: 2427-2430.
  20. Ma, K. L., X. Z. Ruan, S. H. Powis, Y. Chen, J. F. Moorhead, and Z. Varghese. 2007. Sirolimus modifies cholesterol homeostasis in hepatic cells: a potential molecular mechanism for sirolimus-associated dyslipidemia. Transplantation 84: 1029-1036.
  21. Vaziri, N. D. 2003. Molecular mechanisms of lipid disorders in nephrotic syndrome. Kidney Int 63: 1964-1976.
  22. Vaziri, N. D., T. Sato, and K. Liang. 2003. Molecular mechanisms of altered cholesterol metabolism in rats with spontaneous focal glomerulosclerosis. Kidney Int 63: 1756-1763.
  23. Vaziri, N. D., K. Liang, and J. S. Parks. 2001. Acquired lecithin-cholesterol acyltransferase deficiency in nephrotic syndrome. Am J Physiol Renal Physiol 280: F823-828.
  24. Vaziri, N. D., C. H. Kim, D. Phan, S. Kim, and K. Liang. 2004. Up-regulation of hepatic Acyl CoA: Diacylglycerol acyltransferase-1 (DGAT-1) expression in nephrotic syndrome. Kidney Int 66: 262-267.
  25. . 2002. MRC/BHF Heart Protection Study of cholesterol lowering with simvastatin in 20,536 high-risk individuals: a randomised placebo-controlled trial. Lancet 360: 7-22.
  26. Palmer, S. C., S. D. Navaneethan, J. C. Craig, D. W. Johnson, V. Perkovic, J. Hegbrant, and G. F. Strippoli. 2014. HMG CoA reductase inhibitors (statins) for people with chronic kidney disease not requiring dialysis. Cochrane Database Syst Rev 5: CD007784.
  27. Toto, R. D., S. M. Grundy, and G. L. Vega. 2000. Pravastatin treatment of very low density, intermediate density and low density lipoproteins in hypercholesterolemia and combined hyperlipidemia secondary to the nephrotic syndrome. Am J Nephrol 20: 12-17.
  28. Matzkies, F. K., U. Bahner, M. Teschner, H. Hohage, A. Heidland, and R. M. Schaefer. 1999. Efficiency of 1-year treatment with fluvastatin in hyperlipidemic patients with nephrotic syndrome. Am J Nephrol 19: 492-494.
  29. Asselbergs, F. W., G. F. Diercks, H. L. Hillege, A. J. van Boven, W. M. Janssen, A. A. Voors, D. de Zeeuw, P. E. de Jong, D. J. van Veldhuisen, W. H. van Gilst, R. Prevention of, and I. Vascular Endstage Disease Intervention Trial. 2004. Effects of fosinopril and pravastatin on cardiovascular events in subjects with microalbuminuria. Circulation 110: 2809-2816.
  30. Scheven, L., M. Van der Velde, H. J. Lambers Heerspink, P. E. De Jong, and R. T. Gansevoort. 2013. Isolated microalbuminuria indicates a poor medical prognosis. Nephrol Dial Transplant 28: 1794-1801.
  31. Wanner, C., V. Krane, W. Marz, M. Olschewski, J. F. Mann, G. Ruf, and E. Ritz. 2005. Atorvastatin in patients with type 2 diabetes mellitus undergoing hemodialysis. N Engl J Med 353: 238-248.
  32. Fellstrom, B. C., A. G. Jardine, R. E. Schmieder, H. Holdaas, K. Bannister, J. Beutler, D. W. Chae, A. Chevaile, S. M. Cobbe, C. Gronhagen-Riska, J. J. De Lima, R. Lins, G. Mayer, A. W. McMahon, H. H. Parving, G. Remuzzi, O. Samuelsson, S. Sonkodi, D. Sci, G. Suleymanlar, D. Tsakiris, V. Tesar, V. Todorov, A. Wiecek, R. P. Wuthrich, M. Gottlow, E. Johnsson, and F. Zannad. 2009. Rosuvastatin and cardiovascular events in patients undergoing hemodialysis. N Engl J Med 360: 1395-1407.
  33. Baigent, C., M. J. Landray, C. Reith, J. Emberson, D. C. Wheeler, C. Tomson, C. Wanner, V. Krane, A. Cass, J. Craig, B. Neal, L. Jiang, L. S. Hooi, A. Levin, L. Agodoa, M. Gaziano, B. Kasiske, R. Walker, Z. A. Massy, B. Feldt-Rasmussen, U. Krairittichai, V. Ophascharoensuk, B. Fellstrom, H. Holdaas, V. Tesar, A. Wiecek, D. Grobbee, D. de Zeeuw, C. Gronhagen-Riska, T. Dasgupta, D. Lewis, W. Herrington, M. Mafham, W. Majoni, K. Wallendszus, R. Grimm, T. Pedersen, J. Tobert, J. Armitage, A. Baxter, C. Bray, Y. Chen, Z. Chen, M. Hill, C. Knott, S. Parish, D. Simpson, P. Sleight, A. Young, and R. Collins. 2011. The effects of lowering LDL cholesterol with simvastatin plus ezetimibe in patients with chronic kidney disease (Study of Heart and Renal Protection): a randomised placebo-controlled trial. Lancet 377: 2181-2192.
  34. Palmer, S. C., S. D. Navaneethan, J. C. Craig, D. W. Johnson, V. Perkovic, S. U. Nigwekar, J. Hegbrant, and G. F. Strippoli. 2013. HMG CoA reductase inhibitors (statins) for dialysis patients. Cochrane Database Syst Rev 9: CD004289.
  35. Marz, W., B. Genser, C. Drechsler, V. Krane, T. B. Grammer, E. Ritz, T. Stojakovic, H. Scharnagl, K. Winkler, I. Holme, H. Holdaas, C. Wanner, D. German, and I. Dialysis Study. 2011. Atorvastatin and low-density lipoprotein cholesterol in type 2 diabetes mellitus patients on hemodialysis. Clin J Am Soc Nephrol 6: 1316-1325.
  36. Vaziri, N. D., and K. C. Norris. 2013. Reasons for the lack of salutary effects of cholesterol-lowering interventions in end-stage renal disease populations. Blood purification 35: 31-36.
  37. Holdaas, H., B. Fellstrom, A. G. Jardine, I. Holme, G. Nyberg, P. Fauchald, C. Gronhagen-Riska, S. Madsen, H. H. Neumayer, E. Cole, B. Maes, P. Ambuhl, A. G. Olsson, A. Hartmann, D. O. Solbu, and T. R. Pedersen. 2003. Effect of fluvastatin on cardiac outcomes in renal transplant recipients: a multicentre, randomised, placebo-controlled trial. Lancet 361: 2024-2031.
  38. Jardine, A. G., H. Holdaas, B. Fellstrom, E. Cole, G. Nyberg, C. Gronhagen-Riska, S. Madsen, H. H. Neumayer, B. Maes, P. Ambuhl, A. G. Olsson, I. Holme, P. Fauchald, C. Gimpelwicz, T. R. Pedersen, and A. S. Investigators. 2004. fluvastatin prevents cardiac death and myocardial infarction in renal transplant recipients: post-hoc subgroup analyses of the ALERT Study. Am J Transplant 4: 988-995.
  39. Holdaas, H., B. Fellstrom, A. G. Jardine, G. Nyberg, C. Gronhagen-Riska, S. Madsen, H. H. Neumayer, E. Cole, B. Maes, P. Ambuhl, J. O. Logan, B. Staffler, C. Gimpelewicz, and A. S. Group. 2005. Beneficial effect of early initiation of lipid-lowering therapy following renal transplantation. Nephrol Dial Transplant 20: 974-980.
  40. Fellstrom, B., H. Holdaas, A. G. Jardine, I. Holme, G. Nyberg, P. Fauchald, C. Gronhagen-Riska, S. Madsen, H. H. Neumayer, E. Cole, B. Maes, P. Ambuhl, A. G. Olsson, A. Hartmann, J. O. Logan, T. R. Pedersen, and I. Assessment of Lescol in Renal Transplantation Study. 2004. Effect of fluvastatin on renal end points in the Assessment of Lescol in Renal Transplant (ALERT) trial. Kidney Int 66: 1549-1555.
  41. Samuelsson, O., H. Mulec, C. Knight-Gibson, P. O. Attman, B. Kron, R. Larsson, L. Weiss, H. Wedel, and P. Alaupovic. 1997. Lipoprotein abnormalities are associated with increased rate of progression of human chronic renal insufficiency. Nephrol Dial Transplant 12: 1908-1915.
  42. Samuelsson, O., P. O. Attman, C. Knight-Gibson, R. Larsson, H. Mulec, L. Weiss, and P. Alaupovic. 1998. Complex apolipoprotein B-containing lipoprotein particles are associated with a higher rate of progression of human chronic renal insufficiency. J Am Soc Nephrol 9: 1482-1488.
  43. Sato, H., S. Suzuki, H. Kobayashi, S. Ogino, A. Inomata, and M. Arakawa. 1991. Immunohistological localization of apolipoproteins in the glomeruli in renal disease: specifically apoB and apoE. Clin Nephrol 36: 127-133.
  44. Fried, L. F., T. J. Orchard, and B. L. Kasiske. 2001. Effect of lipid reduction on the progression of renal disease: a meta-analysis. Kidney Int 59: 260-269.
  45. Rahman, M., W. Yang, S. Akkina, A. Alper, A. H. Anderson, L. J. Appel, J. He, D. S. Raj, J. Schelling, L. Strauss, V. Teal, D. J. Rader, and C. S. Investigators. 2014. Relation of serum lipids and lipoproteins with progression of CKD: The CRIC study. Clin J Am Soc Nephrol 9: 1190-1198.
  46. Dormuth, C. R., B. R. Hemmelgarn, J. M. Paterson, M. T. James, G. F. Teare, C. B. Raymond, J. P. Lafrance, A. Levy, A. X. Garg, P. Ernst, and S. Canadian Network for Observational Drug Effect. 2013. Use of high potency statins and rates of admission for acute kidney injury: multicenter, retrospective observational analysis of administrative databases. BMJ 346: f880.
  47. Bangalore, S., R. Fayyad, G. K. Hovingh, R. Laskey, L. Vogt, D. A. DeMicco, D. D. Waters, C. Treating to New Targets Steering, and Investigators. 2014. Statin and the risk of renal-related serious adverse events: Analysis from the IDEAL, TNT, CARDS, ASPEN, SPARCL, and other placebo-controlled trials. Am J Cardiol 113: 2018-2020.
  48. Lee, J. M., J. Park, K. H. Jeon, J. H. Jung, S. E. Lee, J. K. Han, H. L. Kim, H. M. Yang, K. W. Park, H. J. Kang, B. K. Koo, S. H. Jo, and H. S. Kim. 2014. Efficacy of short-term high-dose statin pretreatment in prevention of contrast-induced acute kidney injury: updated study-level meta-analysis of 13 randomized controlled trials. PLoS One 9: e111397.
  49. Han, Y., G. Zhu, L. Han, F. Hou, W. Huang, H. Liu, J. Gan, T. Jiang, X. Li, W. Wang, S. Ding, S. Jia, W. Shen, D. Wang, L. Sun, J. Qiu, X. Wang, Y. Li, J. Deng, J. Li, K. Xu, B. Xu, R. Mehran, and Y. Huo. 2014. Short-term rosuvastatin therapy for prevention of contrast-induced acute kidney injury in patients with diabetes and chronic kidney disease. J Am Coll Cardiol 63: 62-70.
  50. Sattar, N., D. Preiss, H. M. Murray, P. Welsh, B. M. Buckley, A. J. de Craen, S. R. Seshasai, J. J. McMurray, D. J. Freeman, J. W. Jukema, P. W. Macfarlane, C. J. Packard, D. J. Stott, R. G. Westendorp, J. Shepherd, B. R. Davis, S. L. Pressel, R. Marchioli, R. M. Marfisi, A. P. Maggioni, L. Tavazzi, G. Tognoni, J. Kjekshus, T. R. Pedersen, T. J. Cook, A. M. Gotto, M. B. Clearfield, J. R. Downs, H. Nakamura, Y. Ohashi, K. Mizuno, K. K. Ray, and I. Ford. 2010. Statins and risk of incident diabetes: a collaborative meta-analysis of randomised statin trials. Lancet 375: 735-742.
  51. Preiss, D., S. R. Seshasai, P. Welsh, S. A. Murphy, J. E. Ho, D. D. Waters, D. A. DeMicco, P. Barter, C. P. Cannon, M. S. Sabatine, E. Braunwald, J. J. Kastelein, J. A. de Lemos, M. A. Blazing, T. R. Pedersen, M. J. Tikkanen, N. Sattar, and K. K. Ray. 2011. Risk of incident diabetes with intensive-dose compared with moderate-dose statin therapy: a meta-analysis. JAMA 305: 2556-2564.
  52. He, Y. M., L. Feng, D. M. Huo, Z. H. Yang, and Y. H. Liao. 2014. Benefits and harm of niacin and its analog for renal dialysis patients: a systematic review and meta-analysis. Int Urol Nephrol 46: 433-442.
  53. Cho, K. H., H. J. Kim, V. S. Kamanna, and N. D. Vaziri. 2010. Niacin improves renal lipid metabolism and slows progression in chronic kidney disease. Biochim Biophys Acta 1800: 6-15.
  54. Charnow, J. A. 2014. Niacin may slow chronic kidney disease progression. In Renal & Urology News.
  55. Kang, H. L., D. Y. Kim, S. M. Lee, K. H. Kim, S. H. Han, H. K. Nam, K. H. Kim, S. E. Kim, Y. K. Son, and W. S. An. 2013. Effect of low-dose niacin on dyslipidemia, serum phophorus levels and adverse effects in patients with chornic kidney disease. Kidney Res Clin Pract 32: 21-26.
  56. Maccubbin, D., D. Tipping, O. Kuznetsova, W. A. Hanlon, and A. G. Bostom. 2010. Hypophosphatemic effect of niacin in patients without renal failure: a randomized trial. Clin J Am Soc Nephrol 5: 582-589.
  57. Maccubbin, D., H. E. Bays, A. G. Olsson, V. Elinoff, A. Elis, Y. Mitchel, W. Sirah, A. Betteridge, R. Reyes, Q. Yu, O. Kuznetsova, C. M. Sisk, R. C. Pasternak, and J. F. Paolini. 2008. Lipid-modifying efficacy and tolerability of extended-release niacin/laropiprant in patients with primary hypercholesterolaemia or mixed dyslipidaemia. International journal of clinical practice 62: 1959-1970.
  58. Boden, W. E., J. L. Probstfield, T. Anderson, B. R. Chaitman, P. Desvignes-Nickens, K. Koprowicz, R. McBride, K. Teo, and W. Weintraub. 2011. Niacin in patients with low HDL cholesterol levels receiving intensive statin therapy. N Engl J Med 365: 2255-2267.
  59. Group, H. T. C., M. J. Landray, R. Haynes, J. C. Hopewell, S. Parish, T. Aung, J. Tomson, K. Wallendszus, M. Craig, L. Jiang, R. Collins, and J. Armitage. 2014. Effects of extended-release niacin with laropiprant in high-risk patients. N Engl J Med 371: 203-212.
  60. Sica, D. A. 2009. Fibrate therapy and renal function. Curr Atheroscler Rep 11: 338-342.
  61. Jun, M., B. Zhu, M. Tonelli, M. J. Jardine, A. Patel, B. Neal, T. Liyanage, A. Keech, A. Cass, and V. Perkovic. 2012. Effects of fibrates in kidney disease: a systematic review and meta-analysis. J Am Coll Cardiol 60: 2061-2071.
  62. Davis, T. M., R. Ting, J. D. Best, M. W. Donoghoe, P. L. Drury, D. R. Sullivan, A. J. Jenkins, R. L. O'Connell, M. J. Whiting, P. P. Glasziou, R. J. Simes, Y. A. Kesaniemi, V. J. Gebski, R. S. Scott, A. C. Keech, I. Fenofibrate, and i. Event Lowering in Diabetes Study. 2011. Effects of fenofibrate on renal function in patients with type 2 diabetes mellitus: the Fenofibrate Intervention and Event Lowering in Diabetes (FIELD) Study. Diabetologia 54: 280-290.
  63. Tonelli, M., D. Collins, S. Robins, H. Bloomfield, and G. C. Curhan. 2004. Gemfibrozil for secondary prevention of cardiovascular events in mild to moderate chronic renal insufficiency. Kidney Int 66: 1123-1130.
  64. Ting, R. D., A. C. Keech, P. L. Drury, M. W. Donoghoe, J. Hedley, A. J. Jenkins, T. M. Davis, S. Lehto, D. Celermajer, R. J. Simes, K. Rajamani, and K. Stanton. 2012. Benefits and safety of long-term fenofibrate therapy in people with type 2 diabetes and renal impairment: the FIELD Study. Diabetes Care 35: 218-225.
  65. Kumbhani, D. 2014. Trial Summary: IMPROVE-IT. In CardioSource.
  66. Haynes, R., D. Lewis, J. Emberson, C. Reith, L. Agodoa, A. Cass, J. C. Craig, D. de Zeeuw, B. Feldt-Rasmussen, B. Fellstrom, A. Levin, D. C. Wheeler, R. Walker, W. G. Herrington, C. Baigent, M. J. Landray, S. C. Group, and S. C. Group. 2014. Effects of lowering LDL cholesterol on progression of kidney disease. J Am Soc Nephrol 25: 1825-1833.
  67. Morita, T., S. Morimoto, C. Nakano, R. Kubo, Y. Okuno, M. Seo, K. Someya, M. Nakahigashi, H. Ueda, N. Toyoda, M. Kusabe, F. Jo, N. Takahashi, T. Iwasaka, and I. Shiojima. 2014. Renal and vascular protective effects of ezetimibe in chronic kidney disease. Internal medicine 53: 307-314.
  68. Rizos, E. C., E. E. Ntzani, E. Bika, M. S. Kostapanos, and M. S. Elisaf. 2012. Association between omega-3 fatty acid supplementation and risk of major cardiovascular disease events: a systematic review and meta-analysis. JAMA 308: 1024-1033.
  69. Mori, T. A., V. Burke, I. Puddey, A. Irish, C. A. Cowpland, L. Beilin, G. Dogra, and G. F. Watts. 2009. The effects of [omega]3 fatty acids and coenzyme Q10 on blood pressure and heart rate in chronic kidney disease: a randomized controlled trial. J Hypertens 27: 1863-1872.
  70. Gopinath, B., D. C. Harris, V. M. Flood, G. Burlutsky, and P. Mitchell. 2011. Consumption of long-chain n-3 PUFA, alpha-linolenic acid and fish is associated with the prevalence of chronic kidney disease. The British journal of nutrition 105: 1361-1368.
  71. Lok, C. E., L. Moist, B. R. Hemmelgarn, M. Tonelli, M. A. Vazquez, M. Dorval, M. Oliver, S. Donnelly, M. Allon, and K. Stanley. 2012. Effect of fish oil supplementation on graft patency and cardiovascular events among patients with new synthetic arteriovenous hemodialysis grafts: a randomized controlled trial. Jama 307: 1809-1816.
  72. Svensson, M., E. B. Schmidt, K. A. Jorgensen, and J. H. Christensen. 2006. N-3 fatty acids as secondary prevention against cardiovascular events in patients who undergo chronic hemodialysis: a randomized, placebo-controlled intervention trial. Clin J Am Soc Nephrol 1: 780-786.

 

 

Dietary Treatment of Obesity

1. Introduction

NHANES data reveals that 33% of U.S. adults are overweight, (BMI of 25-29), over 35% are obese (BMI 30 or higher) and over 6% are extremely obese (BMI greater than or equal to 40.0) (1). Obesity is a chronic medical condition requiring long-term therapy (2, 3-5). If left untreated, overweight and obesity can increase the risk degenerative diseases such as diabetes, hypertension, dyslipidemia, coronary artery disease, and metabolic syndrome, and orthopedic problems. In addition, obesity can promote the development of various negative psychological effects, and can diminish one’s quality of life (6).

Self-initiated approaches to weight reduction are often ineffective. We all long for a quick and easy remedy to cure it, when in fact, there is no sure cure. The only effective method to keep off excess weight is through life-long weight management and obesity prevention, involving physical activity, balanced with a healthy diet (3;5). Health professionals can help people become more effective at maintaining a healthy weight, or losing weight when necessary.

Although 1998 NIH guidelines recommend that healthcare professionals advise obese patients to lose weight, the proportion of obese patients who reported being counseled by a healthcare professional has declined since 1994 (7). Yet the fact remains that a modest (10%) weight reduction in obese people is an attainable goal, and often results in clinical improvements of several health-related parameters, even if the individual remains clinically obese (3;8;9). This information should encourage health professionals to advise weight loss for obese patients and that they need not be overwhelmed by their inability to meet excessively ambitious, or unrealistic, weight loss goals (8;10). Smaller amounts of weight loss can still bring considerable health and social benefits.

There is a great deal of misinformation about obesity in many countries today, including the USA. According to a survey by the Natural Marketing Institute, 59% of the general population would benefit from losing weight. Of them, 26% used weight loss products in the past year, 21% used prescriptions, 18% used over the counter medications and 11% used weight loss dietary supplements to maintain and/or manage their weight (11). Only some of these strategies are effective, as we will see in this chapter.

Weight management counseling of overweight and obese patients deserves reconsideration and reemphasis by health professionals because it carries potential for health benefits. Obese patients receiving weight reduction advice from their physicians are significantly more likely to embark on weight loss attempts than those who do not. Yet less than 42% of obese individuals reported that they received weight loss recommendations from their physicians (12). These findings underscore the need for increased health professional involvement in obesity treatment (4;10;13). When physicians are appropriately aware of, and include recommendations for lifestyle changes in counseling their obese patients, results are promising (12;14). Even more importantly, they should stress achievement and maintenance of a healthy weight before obesity becomes apparent.

The 2010 Dietary Guidelines for Americans (Table 1) provides assistance in maintaining and achieving a healthy weight as well as reducing risk of chronic, diet-related diseases through promoting health and a healthy eating pattern (15). MyPlate.gov (Figure 1) and the Dietary Approaches to Stop Hypertension (DASH) recommendations (Table 2) also provide specific guidance on food selection to assure a healthful diet. The 2008 Physical Activity Guidelines for Americans provide science-based guidance to help Americans, ages six and older, improve their health through appropriate forms of physical activity (Table 3). These guidelines are all useful for prevention of weight gain and maintenance of a healthy weight for the weight loss phase of weight control, different physical activity recommendations are involved, and these are discussed in this chapter.

This chapter focuses on steps health professionals can take to help their patients manage their weights more effectively, or to lose weight by dietary means when that is necessary.

Table 1. Dietary Guidelines for Americans, 2010 (15)
Risk Intervention and Goals Key Recommendations Special Population Recommendations
Adequate nutrients within calorie needs Consume a variety of nutrient-dense foods/beverages with the basic food groupsLimit intake of saturated and trans fats, cholesterol, added sugars, salt, and alcohol.Balance intake of calories with energy needs Adults age >50 should consume vitamin B12 fortified foods or a supplement in crystalline formWomen of childbearing age who may become pregnant should consume foods rich in heme-iron and/or iron-rich plant foods with food rich in vitamin C to enhance absorptionWomen in the first trimester should also consume adequate folic acid via dietary supplement, fortified sources of food containing folic acid as well as other foods naturally high in folic acidOlder adults, people with dark skin, and those not exposed to sufficient sunlight should consume extra vitamin D from vitamin D fortified foods and/or supplements
Weight Management Maintain body weight in a healthy range by balancing calories with energy expendedPrevent gradual weight gain by making small decreases in food/beverage calories in combination with increases in physical activity Overweight adults: strive for slow, steady weight loss by decreasing calories and increasing physical activity while maintaining adequate nutrient intakeOverweight children place on a weight-reduction diet only after consultation with a healthcare provider; reduce the rate of weight gain while allowing growth and developmentPregnant women ensure weight gain is appropriate as specified by healthcare provider, since optimal total pregnancy weight gain varies from person to person (14).Breastfeeding women: moderate weight loss is acceptable and does not compromise of the nursing infant’s weight gainOverweight adults and children with chronic diseases and/or on medications: consult a healthcare provider before starting weight reduction to obtain a weight loss plan that ensures other health problems are managed appropriately.
Physical Activity Engage in regular physical activity and reduce sedentary activities to promote overall health, psychological well-being and a healthy body weightReduce the risk of chronic diseases in adulthood by engaging in at least 30 minutes of moderate-intensity physical activity on most days of the weekGreater health benefits can be obtained by engaging in physical activity that is more vigorous or for a longer durationManage unhealthy body weight gain by engaging in about 60 minutes of moderate-to-vigorous intensity activity on most days per week while not exceeding caloric requirementsSustain weight loss by engaging in at least 60-90 minutes of daily moderate-intensity physical activity while not exceeding caloric requirementsAchieve over-all physical fitness by including a variety of exercises (cardiovascular, stretching, resistance, and calisthenics) Children and adolescents: engage in at least 60 minutes of physical activity on most, preferably all, days of the weekPregnant women: engage in 30 minutes or more of moderate intensity physical activity most days of the week if no medical or obstetric complications are present; avoid falls and abdominal traumaBreastfeeding women: there are no adverse effects from acute or regular exerciseOlder adults: participate in regular physical activity to help reduce functional declines associated with age
Food Groups to Encourage Consume a sufficient amount of fruits and vegetables while staying within energy needs; two cups of fruit and 21/2 cups of vegetables per day are recommended for a reference 2,000-calorie intake, with higher or lower amounts depending on the calorie levelChoose a variety of fruits and vegetables each day. In particular, select from all five vegetable subgroups (dark green, orange, legumes, starchy vegetables, and other vegetables) several times a weekConsume 3 or more ounce-equivalents of whole-grain products per day, with the rest of the recommended grains coming from enriched or whole-grain products. In general, at least half the grains should come from whole grainsConsume 3 cups per day of fat-free or low-fat milk or equivalent milk products. Children and adolescents: consume whole-grain products often; at least half the grains should be whole grainsChildren 2 to 8 years: consume 2 cups per day of fat-free or low-fat milk or equivalent milk productsChildren 9 years of age and older: consume 3 cups per day of fat-free or low-fat milk or equivalent milk products
Fats Consume less than 10% of calories from saturated fat and less than 300 mg/day of cholesterolLimit intake of fats and oils high in saturated and trans-fatty acids; keep trans-fatty acid intake as low as possibleConsume 20% - 35% of total calories from fat; emphasize polyunsaturated and monounsaturated fatty acids (i.e. fish, nuts, vegetable oils)Choose and prepare meat, poultry, dry beans and milk or milk-products that are lean, low-fat or fat-free Children ages 2 to 3 : keep total fat intake between 30%-35% of total caloriesChildren and adolescents ages 4 to 18 : consume 25% - 35% of total calories from fat with most fats coming from sources of polyunsaturated and monounsaturated fatty acids (i.e. fish, nuts and vegetable oils)
Carbohydrates Choose fiber-rich fruits, vegetables, and whole grains oftenChoose and prepare foods and beverages with little added sugars or caloric sweeteners, such as amounts suggested by the USDA Food Guide and the DASH Eating PlanReduce the incidence of dental caries by practicing good oral hygiene and consuming sugar - and starch-containing foods and beverages less frequently  
Sodium and Potassium Consume potassium-rich foods daily such as fruits and vegetables while choosing and preparing foods with little salt Consume less than 2,300 mg (about 1 tsp) of sodium per day
Middle-aged and older adults, African Americans, and people with hypertension aim to consume less than 1,500 mg of sodium per day and meet potassium recommendation of 4,700 mg per day with food.
Alcohol Those who choose to drink alcohol should do so in moderation; defined as 1 drink per day for women; 2 drinks per day for men Those engaging in activities that require attention, skill or coordination such as driving or operating machinery: avoid alcohol consumptionThose under the legal drinking age and women who are pregnant should avoid alcohol consumption.
Food Safety To avoid microbial foodborne illness, clean hands, food contact surfaces, and fruits and vegetablesMeat and poultry should not be washed or rinsedSeparate raw, cooked, and ready-to-eat foods while shopping, preparing, or storing foodsCook foods to a safe temperature to kill microorganismsChill (refrigerate) perishable food promptly and defrost foods properlyAvoid raw (unpasteurized) milk or any products made from unpasteurized milk, raw or partially cooked eggs or foods containing raw eggs, raw or undercooked meat and poultry, unpasteurized juices, and raw sprouts Infants and young children, pregnant women, older adults, and those who are immunocompromised DO not eat or drink raw (unpasteurized) milk or any products made from unpasteurized milk, raw or partially cooked eggs or foods containing raw eggs, raw or undercooked meat and poultry, raw or undercooked fish or shellfish, unpasteurized juices, and raw sprouts.Pregnant women, older adults, and those who are immunocompromised eat only deli meats and frankfurters that have been reheated to steaming hot.

 

Table 2. Dietary Approaches to Stop Hypertension (DSH) Diet recommendations (17)
The number of daily servings in food group vary depending on caloric needs.
Food Group 1,200 calories 1,400 calories 1,600 calories 1,800 calories 2,000 calories 2,600 calories 3,100 calories Serving Sizes
Grains 4-5 5-6 6 6 6-8 10-11 12-13 1 slice bread1 oz dry cerealb½ cup cooked rice, pasta or cereal b
Vegetables 3-4 3-4 3-4 4-5 4-5 5-6 6 1 cup raw leafy vegetable½ cup cut-up raw or cooked vegetable½ cup vegetable juice
Fruits 3-4 4 4 4-5 4-5 5-6 6 1 medium fruit¼ cup dried fruit½ cup fresh, frozen or canned fruit½ cup fruit juice
Fat-free or low-fat milk and milk products 2-3 2-3 2-3 2-3 2-3 3 3-4 1 cup milk or yogurt1½ oz cheese
Lean meats, poultry and fish 3 or less 3-4 or less 3-4 or less 6 or less 6 or less 6 or less 6-9 1 oz cooked meats, poultry or fish1 egg
Nuts, seeds, and legumes 3 per week 3 per week 3 -4 per week 4 per week 4-5 per week 1 1 1/3 cup or 1½ oz nuts2 Tbsp peanut butter2 Tbsp or ½ oz seeds½ cup cooked legumes (dried beans, peas)
Fats and Oils 1 1 2 2-3 2-3 3 4 1 tsp soft margarine1 tsp vegetable oil1 Tbsp mayonnaise1 Tbsp salad dressing
Sweets and sugars 3 or less per week 3 or less per week 3 or less per week 5 or less per week 5 or less per week Less than 2 Less than 2 1 Tbsp sugar1 Tbsp jelly or jam½ cup sorbet, gelatin dessert1 cup lemonade
Maximum sodium limitd 2,300 mg/day 2,300 mg/day 2,300 mg/day 2,300 mg/day 2,300 mg/day 2,300 mg/day 2,300 mg/day  
Footnotes dietary approaches to Stop Hypertension (DSH) Diet recommendations
  1. Eating patterns from 1,200 to 1,800 calories meet the nutritional needs of children 4 to 8 years old. Patterns from 1,600 to 3,100 calories meet the nutritional needs of children 9 years and older as well as adults.
  2. Serving sizes vary between ½ _cup and 1¼ _cups, depending on cereal type. Check product’s Nutrition Facts label.
Table 3. 2008 Physical Activity Guidelines for Americans (18)
Population/Focus Area Key Guidelines
Children and Adolescents

Children and adolescents” 60 minutes (1 hour) or more of physical activity daily.

  • Aerobic: Most of the 60 or more minutes a day should be either moderate- or vigorous-intensity aerobic physical activity, and should include vigorous-intensity physical activity at least 3 days a week.
  • Muscle-strengthening: As part of their 60 or more minutes of daily physical activity, children and adolescents should include muscle-strengthening physical activity on at least 3 days of the week.
  • Bone-strengthening: As part of their 60 or more minutes of daily physical activity, children and adolescents should include bone-strengthening physical activity on at least 3 days of the week.

Encourage young people to participate in physical activities that are appropriate for their age, that are enjoyable, and that offer variety.

Adults
  • All adults should avoid inactivity. Some physical activity is better than none, and adults who participate in any amount of physical activity gain some health benefits.
  • For substantial health benefits, adults should do at least 150 minutes (2 hours and 30 minutes) a week of moderate-intensity, or 75 minutes (1 hour and 15 minutes) a week of vigorous-intensity aerobic physical activity, or an equivalent combination of moderate- and vigorous intensity aerobic activity. Aerobic activity should be performed in episodes of at least 10 minutes, and preferably, it should be spread throughout the week.
  • For additional and more extensive health benefits, adults should increase their aerobic physical activity to 300 minutes (5 hours) a week of moderate intensity, or 150 minutes a week of vigorous intensity aerobic physical activity, or an equivalent combination of moderate- and vigorous-intensity activity. Additional health benefits are gained by engaging in physical activity beyond this amount.
  • Adults should also do muscle-strengthening activities that are moderate or high intensity and involve all major muscle groups on 2 or more days a week, as these activities provide additional health benefits.
Older Adults

The Key Guidelines for Adults also apply to older adults. In addition, the following Guidelines are for older adults:

  • When older adults cannot complete 150 minutes of moderate-intensity aerobic activity a week because of chronic conditions, they should be as physically active as their abilities and conditions allow.
  • Older adults should include exercises that maintain or improve balance if they are at risk of falling.
  • Older adults should determine their level of effort for physical activity relative to their level of fitness.
  • Older adults with chronic conditions should understand whether and how their conditions affect their ability to do regular physical activity safely.
Safe Physical Activity
  • To perform physical activity safely and reduce the risk of injuries and other adverse events, people should:
  • Understand the risks but remember that physical activity is safe for almost everyone.
  • Choose types of physical activity that are appropriate for their current fitness level and health goals, because some activities are safer than others.
  • Increase physical activity gradually over time when more activity is necessary to meet guidelines or health goals. Inactive people should “start low and go slow,” gradually increasing how often and how long activities are done.
  • Protection: using appropriate gear and sports equipment, looking for safe environments, following rules and policies, and making sensible choices about when, where, and how to be active.
  • Those with chronic conditions or symptoms should be under the care of a health-care provider and should consult the health-care provider about the types and amounts of activity appropriate for them.
Women During Pregnancy and the Postpartum Period
  • Healthy women who are not already highly active or doing vigorous-intensity activity should complete at least 150 minutes of moderate-intensity aerobic activity a week during pregnancy and the postpartum period. Preferably, this activity should be spread throughout the week.
  • Pregnant women who habitually engage in vigorous-intensity aerobic activity or who are highly active can continue physical activity during pregnancy and the postpartum period, provided that they remain healthy and discuss with their health-care provider how and when activity should be adjusted over time.
Adults with Disabilities
  • Adults with disabilities, who are able to do so, should complete at least 150 minutes a week of moderate-intensity, or 75 minutes a week of vigorous-intensity aerobic activity, or an equivalent combination of moderate- and vigorous-intensity aerobic activity. Aerobic activity should be performed in episodes of at least 10 minutes, and preferably, it should be spread throughout the week.
  • Adults with disabilities, who are able to do so should also do muscle-strengthening activities of moderate or high intensity that involve all major muscle groups on 2 or more days a week, as these activities provide additional health benefits.
  • When adults with disabilities are not able to meet the Guidelines, they should engage in regular physical activity according to their abilities and should avoid inactivity.
  • Adults with disabilities should consult their health-care providers about the amounts and types of physical activity that are appropriate for their abilities.
Chronic Medical Conditions
  • Adults with chronic conditions can obtain important health benefits from regular physical activity.
  • When adults with chronic conditions perform activity according to their abilities, physical activity is safe.
  • Adults with chronic conditions should be under the care of a health-care provider. People with chronic conditions and symptoms should consult their health-care provider about the types and amounts of activity appropriate for them.

2. RATIONALE FOR DIETARY TREATMENT

Over two-thirds of adults in the United States are currently overweight or obese (19). The percent of overweight and obese people has risen to this point over the past several decades, among men and women, all ethnic groups, all ages, and all education levels. From 1960 to 2010, the prevalence of obesity in the United States more than doubled, going from 13.4% to 36.1% in adults ages 20 to 74, although the prevalence of overweight remained relatively stable (20). In recent years the trend has begun to level off, with one large study finding no significant differences in the incidence of obesity in adults between 2003-2004 and 2011-2012 (21). However, the incidence of both overweight and obesity remain high, and with the medical costs for an obese person averaging $1,429 more per year (in 2008 dollars) than a normal weight person, attention to weight control and maintenance remains paramount for medical practitioners (22).

Weight control has health advantages, and therefore maintaining or achieving a healthy weight is important for all Americans. Obesity is associated with an increase in mortality rates. Obese individuals have an increased risk of death of at least 20% for all-cause and CVD associated mortality (23). Excess weight might contribute to as much as 41% of uterine cancers, and 10% of gallbladder, kidney, liver, and colon cancers (. In weight control prevention it is especially paramount because once weight and adiposity have surpassed healthy levels, they are difficult to reduce. Therefore, it is important for health professionals to monitor the weights of all their patients and to provide anticipatory guidance so that those who are already at healthy weights remain so. The 2010 Dietary Guidelines for Americans (Table 1) stress maintenance of a body weight within a healthy range by balancing calories from foods and beverages with calories expended, by preventing gradual weight gain over time, by making small decreases in foods and beverages, and by increasing physical activity. However, this is easier said than done. The chapter will assist health professionals in operationalizing these recommendations.

3. EVALUATING OVERWEIGHT AND OBESITY

This section outlines a stepwise approach for assessing overweight and obesity.

3.1 Assess Body Fat Burden and Health Status

Before any patient is placed on a reducing diet, where caloric intake is greatly reduced, often alongside increases in energy expenditure, medical assessment of weight, fat distribution, and health risks is essential.

3.2 Measure Body Mass Index (BMI) as an Indirect Measure of Body Fat Burden

Weight should be measured, without clothing, on electronic scales, which provide accurate weights even for heavy patients. Scales should be calibrated to ensure accuracy. Height is best measured with a wall-mounted stadiometer or against a wall rather than on beam-balance scales, which are unsteady and unreliable. Body fat is difficult to measure directly and accurately in clinical practice. Therefore, body mass index (BMI), which is highly correlated with total body fat and future health risks, is recommended as the best surrogate method of capturing body fat, although it can overestimate body fat in individuals with high muscle mass. BMI can be calculated using the following formulas (24):

BMI= (weight lbs ÷ height inches2 ) x 703 or BMI = weight kilograms ÷ height meters2 (24)

Table 4 presents the National Institutes of Health (NIH) classification of BMI values for adults (24). These values are based on abundant data associating higher BMI levels with higher health risks. Although individuals with the same BMI often differ somewhat in the amount of body fat they have, this is still a useful approximation that can be performed quickly and inexpensively in clinical settings.

 

Table 4. Classification of Weight Status by Body Mass Index (BMI)
Classification BMI ( kilogram/m 2 )
Underweight <18.5
Normal weight 18.5-24.9
Overweight 25-29.9
Obesity Class 1 30-34.9
Obesity Class 2 35-39.9
Extreme Obesity Class 3 >40

Individuals with a BMI under 18.5 are classified as underweight, whereas those with a BMI over 25 are considered overweight; those over BMI 30 are classified as class 1 obesity, those over BMI 35 as class 2 obesity, and those over BMI 40 as extreme, or class 3, obesity. In general, the orthopedic and metabolic hazards increase with increasing BMI. Tracking changes in BMI, as well as body weight itself, are easy to use tools for monitoring body composition over time, identifying those at risk for developing overweight and obesity, and monitoring the success of those undergoing weight loss therapy.

The distribution of fat on the body, as well as its sheer amount, also alters risk of some metabolic disorders. The reasons for this are becoming clear as the role of adipose tissue as an endocrine organ is more fully understood. Excess abdominal fat in the viscera, characterized by an accumulation of fat centrally (sometimes referred to as android "apple" or abdominal fat distribution or ectopic fat) is associated with greater risk of certain chronic degenerative diseases than is a peripheral fat deposition pattern (gynoid "pear" or lower body fat pattern).

Although the causal associations between certain diseases and body fat distribution are still a matter of debate (28;27), measuring waist circumference in addition to BMI is still clinically useful in assessing risk posed by body fat distribution (24;5;28-31).

Visceral and subcutaneous fat are difficult to measure accurately in office practice. Waist circumference, taken at the level of the umbilicus (belly button) with a plastic or other type of non-stretchable measuring tape, is a reasonable proxy for assessing the likely size of visceral fat deposits and the extent of abdominal obesity. Waist circumference is easier to measure and more straightforward to interpret than are waist-to-hip ratios. It is generally used as the standard in assessing central vs. peripheral fatness. The cut-points for increased risk are a waist circumference of greater than 35 inches (>88cm) in women, or greater than 40 inches (>102cm) in men (24; 31). Although the usefulness of these absolute cut-offs have been questioned due to the many possible confounding variables in their relationship with health, monitoring changes over time is advocated (32). Measuring waist circumference is most useful for defining risk in obese patients with BMI 25-35 kg/m2. Obese patients with BMIs over 35 kg/m2 already have elevated risk, so waist circumference measurements may be less necessary for them (31).

Table 5 shows how risks of weight related conditions such as type 2 diabetes, hypertension, and cardiovascular disease increase with greater BMI and waist circumference. Patients with high waist circumference may need increased monitoring and treatment of blood pressure, unhealthy blood lipid profiles, and other cardiovascular risk factors. Physical inactivity and smoking increase health risk still further. They act synergistically and apparently increase the severity of the other risk factors present as well as increasing risks themselves in other ways. Elevated serum triglycerides and lower HDL are other markers for increased cardiovascular risk that increase with high waist circumference.

 

Table 5. Classification of Risk of Type 2 Diabetes, Hypertension and Cardiovascular Disease Associated with Weight
Classification of Fatness Status by BMI and Waist Circumference Increase in Disease Risk for Type 2 Diabetes, Hypertension and Cardiovascular Disease Over Normal Weight and Waist Circumference
Waist circumference
Women <35 inches
Men < 40 inches
Waist circumference
Women >35 inches
Men > 40 inches
Underweight (BMI <18.5) --- ---
Normal (BMI (18.5-24.9) --- ---
Overweight (BMI 25-29.9) Increased High
Obese Class 1 (BMI 30-34.9) High Very high
Obese Class 2 (BMI 35-39.9) Very high Very high
Extreme Obesity Class 3 (BMI >40) Extremely high Extremely high

3.4 Document Other Risk Factors and Comorbidities That Increase Risk and Have Other Implications for Therapy

The presence of risk factors or already clinically apparent diseases further increases the health risk of obesity over that evident with high BMI and high waist circumference alone. Table 6 describes different conditions that further add to the adverse health effects of overweight and obesity itself (24). Weight loss can help lower elevated blood pressure, blood glucose, both total and low-density lipoprotein levels (LDL), plasma cholesterol and triglyceride levels, and raise high density lipoprotein (HDL) cholesterol levels in those with abnormally high values. Other modalities of treatment, including pharmacologic therapy, may also be necessary to bring some patients into healthy ranges.

Table 6. Risk Factors and Comorbidities that Increase the Risks of Morbidity from Overweight
Level of Risk Conditions
High Absolute Risk Established coronary heart disease or other atherosclerotic diseaseType 2 diabetesSleep Apnea
High absolute Risk if 3 or More of These Risk Factors are Present HypertensionCigarette smokingHigh low-density lipoprotein cholesterolLow high density lipoprotein cholesterolImpaired fasting glucoseFamily history of early cardiovascular diseaseAge: >45 in men or >55 in women
Increased Risk Increased surgical riskPsychological disorders such as depressionOsteoarthritisHirsutism (presence of excess body and facial hairGallstonesStress incontinenceGynecologic problems such as amenorrhea and menorrhagia

3.5 Determine if the Patient is a Candidate for Weight Loss

All individuals with a BMI over 30, and those with a BMI between 25-29.9 with a high waist circumference or one or more of the risk factors listed in Table 6, are potential candidates for weight reduction. Patients who have a BMI between 25-29.9, but who do not have any risk factors or comorbidities should be counseled to avoid further weight gain (23). The goal of weight control is both the reduction of weight and the maintenance of healthy body weight over the long term. Weight loss should be achieved through a high-intensity lifestyle intervention, which is discussed further in section 4.2, if possible (23). If the patient is not open to weight loss, at least prevention of further weight gain should be attempted. Those with very high BMIs (over 35) are unlikely to be able to achieve sufficient fat loss on a usual low calorie diet of 1,200 to 1,500 calories without regimes that must continue for many months. They should be referred for care to a multidisciplinary team specializing in obesity for treatment with very low calorie diets, and possibly pharmacology or surgery (23).
Some individuals whose weights are at healthy levels and who are without weight associated health problems also may wish to lose weight. These patients need to have their concerns about diet addressed, but should not embark on reducing diets since there are no medical indications for them to do so. They should encouraged to maintain their weight within a healthy range, and counseled to follow dietary recommendations from ChooseMyPlate.gov (Figure 1), the Dietary Approaches to Stop Hypertension (DASH) eating plan (Table 2), or the Dietary Guidelines for Americans (Table 1).

choosemyplate.gov
Figure 1. ChooseMyPlate.gov (F)

 

4. Choose Treatment Options

The following section covers the various means of treating obesity, including dietary changes, medications, and/or surgical options. Diet plays a critical role in all of these options.

4.1 Assess the Patient’s Readiness and Willingness to Lose Weight

The previous sections provide the rationale for assessment of the health risks associated with obesity, the potential health benefits accruing from weight loss, and the importance of then maintaining a healthy body weight. Weight control requires behavioral change, which cannot happen without patient buy-in to the process. Therefore, the health risks of overweight and obesity need to be communicated, and patient readiness to change needs to be established. Table 7 outlines the various stages of behavior change as conceptualized by Kushner based on Prochaska’s model of behavior change, often referred to as the Transtheoretical Model of Behavior Change (6). It is important to note many patients will not progress through the outlined stages linearly, but rather will go back and forth repeatedly among stages. Therefore, timing is important and the clinician must watch for an appropriate time to bring up or follow through on the issue.

Table 7. Transtheoretical Model of Behavior Change (5)
Stage Characteristics Patient Verbal Cues
Pre-contemplation Unaware of problem, no interest in change “I’m not really interested in weight loss. It’s not a problem.”
Contemplation Aware of the problem, beginning to think of changing “I need to lose weight but with all that’s going on in my life right now, I’m not sure if I can.”
Preparation Realizes benefits of making changes and thinking about how to make change “I have to lose weight, and I’m planning to do that.”
Action Actively taking steps toward achieving the behavioral goal, but only for a brief period (less than 6 months) “I’m doing my best. This is harder than I thought.”
Maintenance Initial treatment and behavioral goals reached and sustained for a longer period of time (e.g., more than 6 months) “I’ve learned a lot through this process.”

Often, those who are at highest health risk due to obesity are unaware of how serious their weight-related problems are, or are in deep denial about them. The consequences of excess weight, including long-term implications, must therefore be raised and carefully explained. Helping patients to draw connections between the short-and long-term health consequences of their current weight, and the implications this will have on things they care about, such as their family or the ability to participate in activities they enjoy, may aid in empowering patients to progress through the various stages of behavior change.

Once patient readiness and willingness to lose weight has been established, a plan of attack needs to be jointly devised with the patient. Some patients are ready to start a treatment program immediately, and the patient and counselor are able to begin setting goals together right away. Other patients have reservations or other issues keeping them from reaching the action stage needed to embark upon their weight loss goals, making it important for the counselor to address these road-blocks before moving on. For patients who are not ready to act, the issue should be deferred and brought up again at the next visit, rather than dropping the subject entirely. Some groups of patients are unable or unwilling to embark on a weight reduction program at all. Even patients who are unwilling to embark on a reducing diets may be willing to take steps to avoid further weight gain, or may be willing to work on other risk factors such as smoking cessation or increasing physical activity. These activities should be encouraged. For those who are ready and raring to go, a referral to a registered dietitian should be provided where the subject can be addressed in-depth.

4.2 Decide if Dietary Treatment is the Appropriate Option

Weight reduction with dietary treatment is in order for virtually all patients with a BMI over 30, as well as those with a BMI of 25-29.9 with comorbidities. A dietary approach to weight loss should be executed in the context of comprehensive lifestyle intervention whenever possible. This type of intervention involves frequent, in-person encounters with a trained interventionist in an individual or group setting, and incorporates a moderately reduced calorie diet, increases in physical activity, and the use of behavioral techniques to facilitate adherence to recommendations. The gold standard is a comprehensive, high-intensity, on-site program with greater than 14 sessions in 6 months, provided either in a group or individually, by a trained interventionist, and lasting for at least 1 year. When a comprehensive lifestyle intervention is not feasible, other dietary-based approaches, such as electronically based programs and commercial programs, which will be discussed in further detail later, can be appropriate alternatives (23).

For some patients, however, a low calorie (hypocaloric) diet alone may not be enough to prompt significant and lasting weight loss (34).For patients who have failed to lose on a comprehensive lifestyle program, for those with a BMI greater than 30, or greater than 27 if one or more comorbidities are present, and who are likely to have little success with a purely dietary approach on the basis of a history of many failures, other steps may be in order. This is especially important for those with class 2 (BMI>35) and 3 (BMI > 40) obesity, referral to a multidisciplinary obesity treatment team for adjunctive therapies (i.e., very low calorie diets, pharmacological treatment, and/or gastric bypass surgery) is warranted (23).

4.3 Decide if Drugs will be Useful Adjunctive Therapy to the Reducing Diet

Prescription drugs are one form of adjunctive therapy that may be considered for those with a BMI greater than 30, or a BMI of 27 and above if one or more comorbidities are present, who are unable to lose weight with dietary measures alone. Weight loss drugs are only adjuncts to, rather than substitutes for, reducing diets, however, and a reducing diet will still be necessary. Without a hypocaloric diet, drugs are unlikely to be effective. The addition of weight loss medication to a dietary-based weight loss regimen can help patients lose up to 10% of their initial body weight, with most weight loss occurring in the first six months (35). Table 8 provides an overview of prescription medications that are available. Note that none are totally free of side effects.

 

Table 8. Prescription Medications Available in the United States for Weight Loss (26)
Generic Name + (Trade Name) Food and Drug Administration Approval for Weight Loss Drug Type Common Side Effects
Orlistat
(Prescription: XenicalTM)
*OTC Brand: AlliTM
Yes; long term for adults and children age 12 and older
*AlliTM for adults only
Lipase Inhibitor Gastrointestinal issues (cramping, diarrhea, oily spotting)
Do not take with cyclosporine
Lorcaserin
(BelviqTM)
Yes; long term for adults Serotonin Receptor Antagonist Headache, dizziness, nausea, fatigue, dry mouth
Do not take with Selective Serotonin Reuptake Inhibitors (SSRIs) or Monoamine Oxidase Inhibitors (MAOIs)
Phentermine-Topiramate (QsymiaTM) Yes; long term for adults
Contrindicated in women who are pregnant or may become pregnant
Appetite Suppressant/Seizure Treatment Tingling of hands and feet, trouble sleeping, taste alterations, dry mouth constipation, dizziness, birth defects
Bupropion – Naltrexone
(ContraveTM)
Yes; long term for adults Depression Treatment/Alcohol and Opioid Abuse Treatment Nausea, constipation, headache, vomiting, dizziness, insomnia, dry mouth, diarrhea, increased blood pressure and heart rate, seizures, suicidal thoughts and behaviors
Phentermine
(Adipex-PTM, SuprenzaTM, ZantrylTM)
Yes; short term (up to 12 weeks) for adults Appetite Suppressant Increased blood pressure and heart rate, sleeplessness, nervousness
Diethylpropion
(TenuateTM)
Yes; short term (up to 12 weeks) for adults Appetite Suppressant Dizziness, headache, sleeplessness, nervousness
Phendimetrazine
(Bontril PDMTM, AdipostTM, MelfiatTM)
Yes; short term (up to 12 weeks) for adults Appetite Suppressant Sleeplessness, nervousness
Benzphetamine
(DidrexTM)
Yes; short term (up to 12 weeks) for adults Appetite Suppressant Restlessness, anxiety, sleeplessness, headache
Bupropion
(WellbutrinTM)
No Depression Treatment Dry mouth, insomnia
Topiramate
(TopamaxTM)
No Seizure Treatment Numbness of skin, change in taste
Zonisamide
(ZonegranTM)
No Seizure Treatment Drowsiness, dry mouth, dizziness, headache, nausea
Metformin
(GlucophageTM)
No Diabetes Treatment Weakness, dizziness, metallic taste, nausea
Byetta
(ExenatideTM, BydureonTM)
No Diabetes Treatment Nausea

Many of the Food and Drug Administration (FDA)-approved weight-loss medications are approved only for short-term use (short term is usually interpreted to mean use up to 12 weeks), although some physicians still prescribe them for longer periods of time (35). Only four prescription drugs are currently approved for long-term use in weight reduction: Orlistat (XenicalTM), Lorcaserin (BelviqTM), Phentermine-Topiramate (QsymiaTM), and Bupropion–Naltrexone (ContraveTM).

Orlistat is available for both prescription (XenicalTM) and over-the-counter at a lower dose as AlliTM. Over-the-counter AlliTM is available only to adults aged 18 and older, and is a half-dose version of prescription Orlistat (http://www.myalli.com) (35). Orlistat operates at the level of the gut to inhibit pancreatic lipase, blocking the absorption of about one third of fat consumed. Use over one to two years can lead to a weight loss of five to seven pounds (35). Adherence to a reduced calorie diet with less than 30% calories from fat is necessary while on either Orlistat or Ali. Both Orlistat and Ali’s disadvantages include fat malabsorption, sometimes accompanied by anal leakage, and decreased absorption of fat-soluble vitamins. Because of this decrease in fat-soluble vitamin absorption, patients taking either version of the drug should be advised to take a multivitamin supplement containing fat-soluble vitamins to ensure adequate nutritional status (35). Dietetic counseling is helpful in managing weight loss.

Lorcaserin (BelviqueTM) is another weight loss drug approved for long term use and is available by prescription only. Studies evaluating its effectiveness found that 47% of those who used the drug lost at least 5% of their initial body weight (35). QsymiaTM is another long-term weight loss drug that was approved by the FDA in 2012. QsymiaTM is a combination of an appetite suppressant, phentermine, and a seizure medication, topiramate. Studies found that after 1 year using the recommended dose of the drug, 62% of patients lost greater than 5% of their initial body weight (35). ContraveTM is the newest long-term drug to treat obesity, and was approved in September 2014. ContraveTM is a combination of bupropion, an antidepressant, and naltrexone, a medication used to treat alcohol and opioid dependence. Studies showed that after 1 year, 42% of the non-diabetic patients tested lost at least 5% of their initial body weight. (36). With all long-term weight loss drugs, if at least 5% of initial body weight is not lost by 12 weeks, use of the drug should be discontinued as it is unlikely to be effective later, and therefore the risks outweigh the putative benefits (35).

Phentermine (SuprenzaTM), phendimetrazine (AdipostTM), diethylpropion (TenuateTM), and benzphetamine (DidrexTM) are modestly effective prescribed anorectic agents approved for short-term use (12 weeks in a 12 month period) by the Food and Drug Administration (FDA) (35). Phentermine and diethylpropion are widely prescribed, as they are relatively inexpensive (approximately $30 for a one-month supply), and provide slight stimulatory effects. However, little research has been done on their long-term side-effects (39).

The off-label use of bupropion (WellbutrinTM), a drug originally approved by the FDA for aiding in smoking cessation, has become popular in the past few years for weight control. Bupropion enhances norepinephrine and weakly blocks dopamine reuptake and is being studied for the treatment of obesity. Bupropion could be considered if a patient presenting with obesity wanted to quit smoking as well and lose weight (38). Short term side effects most often reported are agitation, dry mouth, insomnia, headache, nausea, constipation, and tremor. However, its long-term effects on weight loss are not clear, and its use must be accompanied by a low-calorie diet if it is to help in weight loss.

Topiramate (TopamaxTM) and zonisamide (ZonegranTM) are anticonvulsants that were originally approved to treat epilepsy. They are also sometimes used off-label for their weight-loss effects. However, adverse effects have also been reported, most commonly difficulty with memory, parathesia, difficulty concentrating, and mood problems. These drugs are approved by the FDA for epilepsy only, and not for weight loss (38).

Metformin (GlucophageTM) is a diabetes medication that may promote small amounts of weight loss in people with obesity and type 2 diabetes. One study found that patients treated with metformin for diabetes lost 2kg more at 6 months compared to placebo, and maintained at 1 kg at 4 years follow-up. It is unclear, however, if weight loss on metformin is related to improved glucose tolerance or the drug itself (40).

ByettaTM (exenatide) and pramlintide are sometimes used in treating the comorbidities of obesity. Both compounds affect the gastrointestinal hormones that regulate glucose homeostasis, gastric emptying, and satiety. Exenatide (ByettaTM) is used as an adjunctive therapy for improving glycemic control in patients with type 2 diabetes who also take metformin or sulfonylurea. Pramlintide is an adjunctive therapy for patients with type 1 or type 2 diabetes who use insulin at mealtimes. Usually patients with diabetes gain weight with better glucose control, however, with these drugs, better blood glucose control is often associated with weight loss, at least in preliminary studies. The most common side effect of these medications is nausea (38).

Major disappointments have resulted as research on the once promising class of drugs known as cannabinoid (CB1) receptor antagonists has continued. Rimonabant (AcompliaTM) was the first CB1 receptor blocker approved for use in the world. Its suggested use was for patients with a BMI of 30 or more, in conjunction with exercise and diet, to aid in weight loss. CB1 receptors are located in the brain, gastrointestinal tract, adipose tissue, heart, pituitary, and adrenal glands, and if they are stimulated, these receptors increase appetite. Blockage of these receptors is thought to decrease appetite. However, the FDA ruled that Rimonabant carried too much risk to be approved for use in the United States, with side effects including nausea, anxiety, diarrhea, and depressed mood that, in severe cases, led to suicide (38). In 2009, the European Medicines Agency (EMEA) also concluded that the benefits of Rimonabant no longer outweighed the risks, and marketing authorization for the drug in the European Union was officially revoked (43). Investigation into the cannabinoid (CB1) receptor antagonist class of drugs has since ceased (39).

Other areas of research for future weight loss drugs include drugs combining appetite suppressants and those that affect addiction, drugs affecting gut hormones that influence appetite, drugs that work to shrink the blood vessels supplying fat cells, drugs targeting genes associated with obesity, and manipulation of gut bacteria (35).

Sibutramine (MeridiaTM) was a commonly used obesity drug first introduced in 1997. However, it was voluntarily withdrewn from US markets by its manufacturer in 2010 after clinical trial data indicated that the drug increased the risk for heart attack and stroke. It should not be prescribed or used for the treatment of obesity (44).

Some of the surprisingly positive effects with weight loss drugs are due the fact that the medications are not what they seem to be but rather adulterated and contain undeclared drugs. The FDA releases an extensive list of tainted weight loss products, many of which contain undeclared drugs (Table 9) (44). If patients are taking any of these contaminated products, they should be advised to stop immediately. Table 9 provides a comprehensive list of these tainted products on the market from 2011 onwards, along with the undeclared pharmaceutical/chemical included in the product. There are also other drugs that may be added although they are no longer available for distribution through legitimate sources because of adverse and sometimes fatal side effects including Fen-PhenTM, ReduxTM, PondimenTM, fenfluramine, MeridiaTM, and dexfenfluramine.

 

Table 9. FDA’s List of Tainted Weight Loss Products (33)

The Undeclared Drug/Chemical Ingredient is Listed After Each Product in Parentheses

1 Day Diet (Sibutramine) 7 Days Herbal Slim (Sibutramine) 24 Ince (Sibutramine)  
A-Slim 100% Natural Slimming Capsule (Sibutramine) Acai Berry Soft Gel ABC (Sibutramine) Advanced (Sibutramine)
Advanced Blue (Sibutramine) Advanced Slim 5 (SIbutramine) Asset Bee Pollen (Sibutramine)
Asset Bold (Sibutramine) Asset Extreme (Sibutramine) Asset Extreme Plus (SIbutramine)
B-Perfect (Sibutramine) Be Inspired (Sibutramine) Beautiful Slim Body (Sibutramine)
Bella Vi Insane Amp’d/Bella Vi Amp’d Up (Sibutramine) Best Line Suplemento Alimenticio (Sibutramine) Best Share Green Coffee: Brazilian Slimming Coffee (Sibutramine)
Bethel 30 (Sibutramine) Body Beauty 5 Days Slimming Coffee (Sibutramine) Botanical Slimming Soft Gel (Sibutramine)
Burn 7 (Sibutramine) Celerite Slimming Capsules (Sibutramine) Citrus Fit Gold (SIbutramine)
DaiDaiHuaJiaoNang (Sibutramine and Phenolphthalein) Diet Master (Sibutramine) Dr. Mao Slimming Capsules (Sibutramine)
Dr. Ming’s Chinese Capsule (Sibutramine) Dream Body Slimming Capsule (Sibutramine) Extreme Body Slim (Sibutramine)
Fat Zero (Sibutramine and Phenolphthalein) Fruit & Plant Slimming (Sibutramine) Fruit Plant Lossing Fat Capsule (Sibutramine)
Goodliness Fat-Reducing Capsules (Sibutramine) Hot Detox (Sibutramine) Infinity (Sibutramine)
Instant Slim/ Shou Fu Ti Tun Guo Xiang Xing Jian Fei Jiao Nang (Sibutramine) Ja Dera 100% Natural Weight Loss Supplement (Sibutramine) Japan Hokkaido Slimming Weight Loss Pills (Sibutramine, Benzocaine, Phenolphthalein and Diclofenac)
Japan Rapid Weight Loss Diet Pills Green (Phenolphthalein) Japan Rapid Weight Loss Diet Pills Yellow (Sibutramine and Phenolphthalein) Japan Weight Loss Blue (Sibutramine, Analogs of Sibutramine, and Ephedrine Alkaloids)
Jimpness Beauty Fat Loss Capsules (Sibutramine) La Jiao Shou Shen (Sibutramine) Leisure 18 Slimming Coffee (Sibutramine)
Lingzhi Cleansed Slim Tea (Sibutramine) Lipo 8 Burn Slim (Sibutramine) Lishou (Sibutramine)
Lite Fit USA (SIbutramine) Lose Weight Coffee (Sibutramine) LX1 (DMAA)
Magic Slim (Sibutramine) Magic Slim Tea (Sibutramine) Magic Slim Weight Reduction Capsule (Sibutramine)
MAXILOSS Weight Advanced (Sibutramine) MAXILOSS Weight Advanced Blue (Sibutramine) Meizi Evolution (Sibutramine)
Meizitang Citrus (Sibutramine) Mix Fruit Slimming (Sibutramine) Natural Body Solution (Sibutramine)
New You (Phenolphthalein) P57 Hoodia (Sibutramine) Pai You Guo Slim Tea (Sibutramine and Phenolphthalein)
Paiyouji Plus (Sibutramine) Perfect Body Solutions (Sibutramine) PhentraBurn Slimming Capsules (Sibutramine)
Sheng Yuan Fang (Sibutramine) Slender Slim 11 (Sibutramine) Slim Forte Slimming Capsule/Slim Forte Double Power Slimming Capsule (Sibutramine)
Slim Forte Slimming Coffee (Sibutramine) Slim Max (Sibutramine) Slim Trim U (Sibutramine)
Slim Xtreme Herbal Slimming Capsule (Sibutramine) SLIMDIA Revolution (SIbutramine) SlimEasy Herbs Capsule (Sibutramine)
SlimExtra Herbal Capsule (Sibutramine) Slimming Diet (Sibutramine) Slimming Diet Berry Plus (Sibutramine)
Strawberry Balance (Sibutramine) Super Slim (Sibutramine) Super Slimming (Sibutramine)
Sport Burner (Fluoxetine) Tengda (Sibutramine) Thinogenics (SIbutramine)
Toxin Discharged Tea (Fluoxetine) Trim-Fast Slimming Softgel (Sibutramine) Ultimate Formula Bee Pollen Capsules (Sibutramine)
Vitaccino Coffee (Sibutramine) XIYOUJI QINGZHI CAPSULE (Sibutramine) Zi Xiu Tang Bee Pollen Capsules (Sibutramine)

Drugs for weight loss are of limited efficacy, some patients cannot afford them, and all of them have side effects. About one fourth of all individuals who are prescribed medications will not have the expected response (39). Patients who are likely to respond to drugs tend to do so within the first month of therapy. If they fail to lose four pounds (1.8 kilograms) in the first four weeks, the drug is unlikely to be effective, and it may be appropriate to discontinue its use. A loss of four pounds within the first four weeks generally predicts weight loss of at least 5% body weight by six months of therapy, if the diet and drug continue to be used (39).

Dietary supplements purported to be helpful in weight loss are discussed in section 9.7.3 (Dietary Supplements and Weight Loss). No supplement currently on the market is both safe and effective for weight loss.

 

4.4 Rule Surgical Options In or Out

Surgical options such as adjustable gastric banding, or more invasive techniques such as rou en y gastric bypass,, sleeve gastrectomy or biliopancreatic diversion with duodenal switch surgery are recommended only for patients classified as Obesity class 2 or above (BMI>35), or as Obesity class 1 (BMI >30) with comorbidities (23). Patients who opt for the surgical route must adhere to certain dietary recommendations before the surgery is performed to show they are able to follow a hypocaloric diet. After surgery, food intake is altered and meals must be smaller because gastric capacity is considerably limited (46). Patients will be required to adhere to a strict, multi-stage diet post-surgery to heal and adjust to new gut physiology short term, as well as to promote weight loss long-term. A post-operative weight reduction surgery diet used in one hospital is shown in Table 10, but there is no standard, widely accepted protocol for diet therapy post-bypass at present. Dietary restrictions must continue indefinitely after surgery to prevent weight regain, and patients will require lifelong use of appropriate vitamin and mineral supplementation to prevent deficiencies. Failure to adhere to a hypocaloric diet through such strategies as consuming large amounts of alcoholic or sugar sweetened beverages, melted ice cream, and many small but calorically dense meals will result in weight and fat gain.

While data suggests that bariatric surgery is more successful than non-surgical interventions, in promoting greater long-term weight loss and inducing initial remission of type-2 diabetes, there is a lack of evidence assessing the long-term risks, complications, and costs of bariatric surgery (238). It is important to note that post-surgery, patients are at risk for many nutritional deficiencies, which can negatively impact overall health, even in the context of weight loss. In one recent study that followed patients who had lost weight more than 50 lbs prior to body contouring procedures, those that had lost more than 100 lbs were more likely to suffer complications, with the effect being greater in those who had lost over 100 lbs from bariatric surgery compared to non-surgical means. Among patients who had bariatric surgery, the risk was highest for those who had had gastric bypass, and lowest in those who had has a lap-band or gastric sleeve (239).

Table 10. Post Gastric Bypass Surgery Diet Used in Tufts Medical Center (34)
Stage 1 One ounce of water per hour, typically in the hospital on the day of surgery
Stage 2 Non-caloric clear liquids, usually in the hospital the day after surgery (e.g., sugar-free Jell-O, flat diet soda, diet juice)
Stage 3
  1. 3-4 small meals per day, each consisting of a high-protein, no added sugar shake, such as Isopure or Sugar-Free Carnation® Instant Breakfast™
  2. Water or non-caloric, non-carbonated clear liquids between meals
  3. Goals of this stage are to drink 64 oz fluid per day 50-60 grams of protein a day for women and 60-70 grams of protein per day for men
  4. This stage lasts 2-3 weeks
Stage 4
  1. Small portions of moist, ground/pureed foods.
  2. Begin supplementing with a multivitamin plus minerals, Vitamin D with calcium (specifically calcium acetate), and sublingual Vitamin B 12
  3. Aim for 60-70 grams of protein per day
  4. This stage lasts 4-5 weeks
Stage 5
  1. Small portions of low-fat (<3-5 grams per serving) or low-sugar (<14 grams per serving) solid foods
  2. At least 64 ounces of fluid per day
  3. Aim for 60-80 grams of protein
  4. Continue to take supplements
  5. Follow this 6-8 weeks after surgery and follow up with a Registered Dietitian

Note: Post Gastric Bypass Surgery Diet Used in Tufts Medical Center is adapted with permission from Melissa Page, MS, Rd, LDN, Weight and Wellness Center, Tufts Medical Center, Boston, MA.

There are many tools available to practitioners to aid in accessing obesity and its related health risks, as well as in determining appropriate treatment options. One such tool is the American Society of Bariatric Physicians Obesity Algorithm (292). Another useful tool is the algorithm in the “2013 Guideline for the Management of Overweight and Obesity in Adults,” which was created by the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and The Obesity Society (23).

5. Set Goals

Goal setting is an important part of achieving weight loss. This section outlines steps in goal setting that are successful for weight loss/ or weight maintenance.

5.1 Clarify Reasonable Goals

In 1998 the National Institutes of Health (NIH) issued guidelines recommending that healthcare professionals advise obese patients to lose weight. Evaluating the effect of these guidelines somewhat later, one study examined the proportion of obese patients and those that actually received advice between 1994 and 2000. The proportion of patients that were obese and received advice to lose weight from physicians during routine medical check-ups decreased from 42.3% in 1994 to 40.3% in 2000. Among those that did receive advice to lose weight had 2.8 odds of trying to lose the weight opposed to those who did not receive any advice (48). In addition to the NIH guidelines, in 2003, the U.S. Preventative Services Task Force (USPSTF) recommends that clinicians screen all adult patients for obesity and offer intensive multi-component behavioral intervention to those affected individuals. Primary care physicians play a critical role in screening adults for obesity and providing appropriate treatment (49). Even with these national recommendations and guidelines, it is still a challenge for healthcare providers to manage obesity (50). Another challenge is who is to pay for it, since many insurance plans do not, and many of those who are obese do not have the money to pay for treatment out-of-pocket. In 2009, less than 50% of obese patients received weight loss advice from physicians, citing many barriers to counseling and intensive treatment of obesity (50). This study concluded that there is a general lack of consensus about the best approach to weight loss; however, this can be improved if counseling skills are developed early during training of physicians in treating obesity (50).

As demonstrated in the study mentioned above, those patients who received advice to lose weight were almost three times more likely to attempt weight loss, so it is important for health professionals to discuss weight loss with their obese patients. The approach used by health professionals when treating obese patients and helping them to set goals is most effective when it is non-judgmental, respectful, and empathetic. This allows patients to feel comfortable with discussing their weight. The focus should be on an acceptable weight to achieve better health outcomes rather than simply reaching a lower body weight (51-53). Furthermore, health professionals must always keep in mind that overweight or obese patients might be hesitant to broach the topic of weight loss, but indeed want assistance in discussing, setting and achieving weight loss. By speaking and working with patients as partners in reaching their weight goal, health care providers can play a big hand in improving their patients’ health.

Understandably patients do not like the terms “obesity,” “fatness,” or “excess fat,” and are more receptive to terms such as, “weight” and “excess weight.” It may be difficult for health care providers to bring up the topic of weight loss, but for more tips and advice, healthcare providers can visit The National Institutes of Health’s Weight Control Information Network (WIN) at http://win.niddk.nih.gov/publications/talking.htm for more information on discussing obesity with patients and for some examples of how to bring up the topic of weight loss with patients.

Determining whether a patient is motivated to lose weight is pertinent and the first order of business, since patient involvement and investment are essential for weight loss success. If a patient is not ready to engage in weight loss, discuss the importance of weight maintenance and physical activity at visits until the patient is ready to begin weight loss therapy (24). Once the patient expresses the desire to lose weight, determine the patient’s weight-related goals and ascertain that goals are realistic and attainable. Agreeing on realistic goals facilitates maintenance of weight loss (54). Unrealistic goals should be discussed and made into more achievable ones. Additionally, patients need to be reassured that the counselor, or healthcare provider, is interested in their health as individuals. Providers should not assume that all of their obese patients’ health problems are weight-related (55), nor should they fail to treat these other problems even if the patients refuse treatment for the obesity. The health care professional’s job is to reduce health risks and improve quality of life to the greatest extent possible within patients’ wishes (56).

On their own, patients often choose to lose weight using drastic measures, such as fasting or very, very low calorie diets, to get fast results even though there is no evidence that they are the best for losing weight. Healthy weight loss is key for long-term weight maintenance (54;57). Intensive, very low-calorie diets (VLCDs; e.g., ≤800 calories per day and especially <500 calories) produce significantly greater initial weight loss, however, these results are often not maintained over time (58). In a study done in 2008, participants were enrolled in a program designed to help maintain weight loss. Participants all initially lost weight using one of three methods, VLCD, commercial programs or a self-guided approach. At the start of the study, those who had used a VLCD lost up to 24% of highest body weight in the last two year compared to those in the commercial programs and self-guided approaches who lost 17%. Results showed that those who used a VLCD regained significantly more weight than the other two groups by six months in the weight maintenance program. However, those who had lost weight using a self-guided method were able to maintain their initial weight loss with great success (58). It should be emphasized that the primary reason for losing weight is for better health outcomes; therefore, weight loss should occur by using healthy methods, ones that can be maintained throughout life. Concentrating on improving health outcomes and other risk factors rather than simply on weight loss is vital.

Once it is established the patient is motivated, health professionals should begin by working with the patient to set realistic, achievable, and sustainable weight loss goals (57). From the medical perspective, an ideal weight goal is one that will maximize heath related effects while minimizing disruption to the patient’s quality of life. This allows the patient to incorporate dietary changes into daily life and therefore incorporate them into their lifestyle to maintain weight loss. Physicians have access to measurements of weight-related risk factors that will be improved if weight is lost. Therefore they are uniquely qualified to define and communicate what a "healthier" weight should be for the patient. Nurse practitioners, registered dietitians, physician assistants, and others should reinforce the message the physician gives, and carry out the actual therapy.

There is no single target weight that will meet every one’s goals. Optimal weight reduction targets vary depending on the patient’s weight and co-morbidities. Progress toward healthier weight goals should involve a gradual approach that minimizes health risks and is timed to the patient’s level of readiness. Excess emphasis on aesthetic and cosmetic aspects of weight loss should be avoided. Most patients may have unrealistic ideas of how much better they will look with weight loss so it is important to stress the health advantages of even modest weight loss (i.e. a half-pound per week) (59). While some cosmetic improvement is possible with a weight loss of five to ten pounds, it rarely meets the patients’ expectations, which can be discouraging. Patients need to have a realistic weight loss target set for themselves, which they have developed during counseling session with a health professional. In time, greater weight loss may be possible if realistic goals are adopted, met, and sustained. The aesthetic and cosmetic effects of weight loss are "extra benefits." The primary medical concern is to help the patient lose enough weight to improve or maintain his/her health.

 

5.2 Adopt Realistic Goals that Include Health Objectives

A healthy target for loss is usually to achieve a weight loss of one-half to two pounds of body weight each week over six months (24 weeks), leading to a decrease of 5 to 10% in body weight from baseline. For example a 250 pound, 5’6’’ woman with a BMI of 40 that lost 12.5 pounds over six months, would have a 5% weight loss, and an ending BMI of 38.5. A 10% weight loss would result in a BMI of 36.3. The goal is to maintain this weight loss over time, and that is never easy. A weight loss of 5 to 10% is achievable and moderate enough to decrease some obesity-related risk factors, such as type 2 diabetes, hypertension, cardiovascular disease, and sleep apnea (9).

The recommendation of dieting for six months rather than a longer amount of time is a practical one because after about six months, most patients have great difficulty sustaining adherence to any diet, particularly if it is very rigorous. Weight plateaus as energy intake fluctuates and resting metabolic rate and energy output decrease. After six months of weight loss, patients should focus on maintenance of the weight loss through a combination of diet therapy, physical activity, and behavior modification. If successful, after several months they can start a weight loss cycle again. When patients do not engage in a weight management program that includes all three components, the risk they will regain all or some of the weight increases (57;60). Most individuals regain one-third of their lost weight in one year, and nearly half return to their original weight within five years (54). The more frequently a patient has contact with his/her health care provider, the weight loss and maintenance outcomes tend to be more successful (57).

 

5.3 Define Successful Outcomes for Weight Reduction with the Patient

Patient weight goals depend on their motivation and their perception of health risks, in addition to other aspects of their lives unrelated to health outcomes. Some patients simply are not motivated to lose weight, or they may be motivated but unwilling or unable to make any changes at present. It is unreasonable provide an unmotivated patient a weight loss goal that they do not wish to achieve. Instead it would be beneficial to come to an agreement about the steps the patient is willing to take to begin to improve their health. One of the newer techniques used in counseling is “motivational interviewing” (MI); an egalitarian, empathetic approach to counseling. It uses specifics strategies, such as reflective listening, positive affirmation, and agenda setting to engage the patient in health behavior changes that will facilitate weight loss. The goal in MI is to assist individuals to work through their ambivalence about behavior change and to find what motivates them internally. Patients understandably often have strong aversions to the weight loss process, so MI counselors reflect the patient’s doubts and work through those with the patient. The process also provides opportunities for the patient to voice concerns about remaining overweight or gaining even more weight, which might serve as motivation to begin losing weight or gaining more control of their weight. MI can be more effective than the counselor simply stating facts to counter the beliefs and doubts of the patient and often enables patients to develop their own reasons and plans for change. Individuals are more likely to accept and act upon their own choices and opinions when they voice them themselves (30).

For patients who are already motivated, the weight loss process can begin much faster because the patient is already willing to make changes. However, it is important to check what they think they will accomplish and when. Their patients’ weight loss targets are often unrealistically low and their time frames unrealistically short (e.g., targets of 25% or more of body weight in a few weeks rather than many months). Numerous studies have shown that obese individuals hope to lose 25 to 35% of their initial weight within a year or less after beginning obesity treatment. Realistically, patients lose only 5 to 15% of their initial weight over a year after beginning any kind of obesity treatment. Unfortunately, many dieters still maintain unrealistic standards even when they are repeatedly informed that their goals may be unrealistic (61).

For example, in one recent assessment, the before-treatment weight loss goals of 45 obese women were assessed, and the women were randomized into a behaviorally based weight-loss program over 48 weeks. While 8 to 10% of weight loss would have been a success from a medical perspective, the women identified a loss of 32% of their body weight as ideal. At the conclusion of their 48-week treatment, the women lost an average of 16% of their total body weight. Even though their weight loss was more than medically expected, the women collectively considered this loss to be “disappointing” (54). This study illustrated how most patients’ ideal weight loss goals are unrealistic and often two to three times what most patients achieve (62). For these types of patients, counseling on more realistic targets and time frames is helpful.

Because patient weight goals are often very different from those of their healthcare providers, health professionals must clearly understand patient expectations for treatment and understand the rationale behind these patient expectations. It may be necessary to work with the patient to re-evaluate changes in expectations over the course of treatment (54). Providing patients with verbal and written information on how much weight they can expect to lose with obesity treatment is helpful in communicating and setting realistic weight loss goals (61). When patients’ ideals of drastic weight loss are not met within their preferred time frame it leads to disappointment and frustration, so these dramatic goals must be addressed at the onset of treatment. It is also important to praise patients once they begin to make positive behavior changes. It helps them feel as though their efforts have been acknowledged and motivates them to maintain their new habits and continue to lose weight or stop gaining more weight. Health professionals should be sure to frequently remind their patients of the health benefits that a 5 to 10% weight loss will offer.

 

5.4 Define Dieting Success in Broader Terms than Weight Loss Alone

Definitions of success are always patient-specific, but health professionals should emphasize the importance of health outcomes rather than how the patient would like to look as their definition of weight loss success. The definition of successful obesity treatment includes goals other than weight loss, and these broader health goals need to be communicated to patients. The reduction of risk factors and co-morbidities, even if weight is not lost, is a "success" from the health standpoint. For some patients, prevention of further weight gain after years of a slow, steady increase in weight is “progress.” The maintenance of a reduced weight, even if it is still within the range of obesity as clinically defined, is also a "success". Some outcomes to focus on include improved metabolic profiles such as, lower blood pressure, serum cholesterol, or fasting blood glucose. The following health behavior changes also denote success: increased daily physical activity and fitness; greater healthfulness of eating patterns, such as more consumption of fruits, vegetables and fiber; and reduction in dietary fat. Changes in specific unhealthful habits such as smoking, or overindulgence in alcoholic beverages, are also reasonable measures of success that may help enhance self-esteem, self-efficacy, quality of life and functional capacity (24;5).

5.5 Set an Individualized "Healthier Weight" Target with the Patient

Patients can be unreasonably hard on themselves and fear that losing weight requires drastic measures. An initial healthy weight goal of 1-2 BMI units often requires much less extreme measures than patients think. For example, a 5’4’’ woman weighing 250 pounds with a BMI of 43 losing 5% of her body weight, or 12.5 pounds, will have an ending BMI of 41. This amount of weight loss could take up to 25 weeks, if she loses one-half a pound per week. Weight loss of one-half a pound to two pounds per week is reasonable and offers the best chance for long-term success, but for extremely heavy people, this may take many months or years. However, as mentioned before, weight loss of even 10% of initial body weight, if sustained, significantly reduces risks of coronary heart disease and other co-morbidities (9;63). Obese patients often expect to lose 25% to 35% of their initial weight over the first year of obesity treatment. Dieters often maintain these expectations even when they are repeatedly informed that their goals are likely unrealistic —even with pharmacological treatment, so the message needs to be repeated (61).

 

5.6 A Reasonable Target: 10% Loss of Body Weight over 6 Months

A 10% weight loss target can be achieved in most patients with a caloric deficit of 500 to 1,000 calories per day, leading to losses of one pound to two pounds per week. For women, a weight reduction plan of eating approximately 1,000 to 1,200 calories per day is suitable. According to the National Institutes of Health and the National Heart, Lung, and Blood Institute, a 1,200 - 1,600 calorie allowance for men or women who weigh 165 pounds, or more, and who exercise regularly is recommended. These calorie amounts along with increased physical activity and behavioral modification will likely produce a caloric deficit to achieve the targeted weight loss plan of one to two pounds per week (64).With a caloric deficit of 500 to 1000 calories per day, if followed with perfect adherence, after six months, weight loss of 26 to 52 pounds would be expected. However, in reality, losses are usually between 20 to 25 pounds, since adherence is never perfect (65).

A decrease of one BMI unit usually represents a loss of 10 to 15 pounds, but the exact amount depends on height and weight. A decrease in two BMI units over six months is another way of stating a weight loss goal. Reductions of this magnitude in weight usually decrease several weight-related risk factors such as blood glucose and blood pressure, which should result in better overall health. In addition, patients’ clothing is likely to fit better and their appearance should be trimmer. If further weight reduction is necessary after 10% of initial body weight is lost, it can be attempted with an increased calorie deficit after prior weight loss has been maintained for several months. Medical nutrition therapy for obesity should last at least six months or until weight loss goals are achieved. After that it is vital to begin a weight maintenance program that includes the same three components used for initial weight loss, diet, physical activity, and behavior change to help prevent weight regain and maintain the patient’s new, healthy lifestyle (57). In addition, the patient should have a strong social support network of encouraging friends and family, and/ or participate in a group where others are also undergoing weight loss treatment. With a strong social support network it is easier for patients to continue his/her healthier lifestyle (66).

 

5.7 Set an Increased Physical Activity Goal

Physical activity is important in weight loss. Physical activity and exercise are not synonymous. Both are desirable but the first is essential. If left on their own, most dieters become more sedentary during weight loss, especially if diets are very low in calories. This is because a markedly negative energy balance reduces exercise tolerance, the body’s maximal power output and increases the body’s sense of perceived exertion (67). Therefore, conscious efforts to increase physical activity while dieting should be attempted. However, physical activity alone, without a reduction of calories, only induces modest reductions in body weight. Few studies to date have incorporated enough physical activity to achieve even a 5% weight loss using a physical activity intervention alone. When physical activity is paired with energy restriction, it has a synergistic effect on weight loss. Despite its modest effects on weight loss, physical activity is also essential for improving health-related outcomes relevant to many obesity related co-morbidities (e.g., heart disease, type 2 diabetes, and possibly some cancers)(68). Physical activity is also vital in preventing weight regain and may enhance quality of life (69). There is a strong association between physical activity at follow-up and maintenance of weight loss. Data from the National Weight Control Registry, a registry of more than 3,000 individuals who have successfully maintained at least a 30-pound weight loss for a minimum of one year, shows that 90% of the individuals report that physical activity is crucial to their long-term weight maintenance. They report expending, on average, 2,700 calories per week in exercise, the energy equivalent of walking four miles seven days a week (70).

5.8 Individualize the Diet and Treatment Program

Evidence-based reviews of successful weight control techniques increasingly emphasize the importance of individualized, multidisciplinary care in addition to realistic goals that are focused on health-outcomes and making permanent lifestyle changes, including an increase in physical activity (65;71).

The specific factors that induce a chronically positive energy balance differ among individuals. Daily lifestyle, environment, resources, and social situations may vary considerably. Weight loss strategies must, therefore, be individualized in order to promote adherence and success (5). No single diet works for everyone. Different dietary approaches for maximizing adherence are successful to varying degrees in different individuals. If asked, patients can usually identify some strategies that have worked for them in the past and health professionals can build a program starting with these strategies as starting points. Previous pitfalls can also be identified and the new weight loss strategy can be tailored to avoid them. Candidates for weight reduction should discuss the approach that best suits their needs with their physician, dietitian, or other health professional. In addition to energy content, individual food selections, meal frequency and many other factors can be tailored to make the diet better suited for the individual. Some factors to consider include the diet’s cost, convenience, how it approaches treatment of co-existing health conditions, and whether it assists patients in adopting strategies for healthful life-long weight maintenance (72;73).

Many overweight patients have already tried many times to lose weight on their own. For example, in the United States 50 – 70% of US adults are trying to lose weight (74). Self-directed efforts are usually motivated by aesthetic or social rather than health-related reasons. The goals they adopt are often unrealistically ambitious, the information they obtain on weight management is often inaccurate, and the motivation and support they receive is often inadequate. Solo efforts often fail and lead to discouragement and a sense of futility (75). The vital role of health professionals is to provide motivation, information, counseling, and support for patients to be successful.

Throughout weight loss patients must be counseled on sound eating patterns. Some dietary education topics that should be discussed to help them are listed in Table 11. The National Institutes of Health (www.nutrition.gov), the American Dietetic Association (www.eatright.org) and other organizations, provide materials, checklists, guidelines, menus, and recipes to assist in such patient education (4;48). Resources for health professionals and for patients can also be accessed at websites such as myplate.gov, the American Heart Association (http://www.americanheart.org/), American Diabetes Association (http://www.diabetes.org/), the American Cancer Society (http://www.cancer.org/), and the American Dietetic Association (http://www.eatright.org/).

 

Table 11. Checklist of Nutrition Education Topics to Cover in
Counseling Patients on Weight Management
(15:57)

Dietary Interventions

  • Energy values of different foods
  • Food Composition (calories, fats, carbohydrate, fiber, protein)
  • The Dietary Guidelines for Americans
  • Portion control and standard serving size (individualized)
  • Meal planning and food preparation
  • Recipe modification
  • Avoiding over consumption of foods with high energy content but little nutritional value
  • Hydration status and limiting alcohol consumption
  • Discuss health risks of obesity and rapid weight loss (monitor health with a team of healthcare providers)

Physical Activity

  • Increases in physical activity (individualized)

Behavior Change

  • Reading nutrition labels
  • Cooking more meals at home
  • New habits of food purchasing
  • Mealtime strategies to avoid overeating
  • Eating strategies for restaurants and social situations
  • Awareness of physiological hunger and satiety cues
  • Awareness of eating and emotions
  • Establishing achievable short-term and medium-term goals
  • Tracking intake and physical activity to keep accountability and records of progress
  • Seeking support

Patients should maintain daily records of their food and beverage intake; this helps to cue them to restrain themselves, and also the record is a visual reminder that it is important to watch one’s intake. Some may also wish to include their mood at the time they ate in order to help in recognizing reasons for eating beyond hunger. Record keeping often increases awareness of consumption, and promotes dietary adherence. Patients should be encouraged to review food records each week and to identify any patterns related to eating and behavior that they can work on for the next week.

6. Plan the Weight Reduction (Energy Deficit Phase) of Weight Control

This section will cover guidelines for the calorie-deficit phase of weight loss.

6.1 General Principles

For an individual already overweight, successful weight control first requires a hypo-caloric phase during which dietary intake is decreased while energy output is increased (or at least not decreased). This phase is referred to as the ”weight loss” "energy deficit" or "hypo-caloric phase" of weight loss.

The essential components of weight loss, regardless of type of diet, are decreased energy intake, increased energy output through physical activity, behavioral modification and alterations in the environment that foster all of these measures. Although this chapter focuses on dietary measures in the treatment of obesity, all reasonable weight control programs should also include physical activity and behavioral modification.

 

6.2 Size of Caloric Deficit Needed to Lose Weight

Obesity results from the accumulation of excessive body fat, which is stored as adipose tissue. An energy deficit of approximately 3,500 calories is required to lose one pound of fat. However, there are several factors that can influence this particular number. These include compensatory changes in resting metabolism, the energy cost of work, and discretionary physical activity, which can sometimes alter this figure by 100 to 200 calories. Over the long-term, this relationship of 3,500 calories per pound of fat holds up quite well. Thus, it is the size of the energy deficit between basal energy needs and the energy output that determines the slope of decline in adipose tissue over time. How well this energy deficit is maintained throughout the weight loss period is dependent on a multitude of factors (76). In addition, creating a calorie deficit using this rule of 3,500 calories per pound may not be applicable to everyone. Work by Dr. Kevin Hall has shown that initial body fat as well as the magnitude of weight loss can influence the applicability of this rule (76).

As previously mentioned, a reduction of 500 to 1,000 calories per day is recommended to achieve a weight loss of approximately one to two pounds of body weight per week (i.e. -3,500 to -7,000 calories total). Cutting down on alcohol, dietary fats and/or sugary caloric carbohydrates is a practical way to produce this deficit (70).

 

6.3 Goal of the Energy Deficit Phase

The goal of dietary treatment of obesity during the weight loss (energy deficit) phase is to decrease body fat stores without unduly depleting lean body mass or otherwise compromising health. Lean body mass includes skeletal muscle and vital organs. During weight loss, some lean muscle tissue is always lost in combination with the fat loss, but the goal is to keep this loss to a minimum (64). While weight is shed, body stores of other nutrients such as water, vitamins, minerals and electrolytes must be maintained. Fortunately, dietary strategies are available to minimize loss of lean tissue and other nutrients.

A systematic review found that increased lean tissue is lost if the energy deficit of the diet is too large in combination with rapid weight loss. In contrast, inclusion of exercise (both cardiovascular and resistance) and adequate dietary protein (60 grams per day, ranging from 0.8 g - 1.5 g per kg of body weight) helps to minimize lean tissue loss (73). These dietary strategies should be incorporated into dietary treatment plans to minimize lean body mass reduction and maximize fat loss.

7. Troubleshoot Diet Failures

There are many factors that contribute to patients losing less weight than expected. This section describes how health professionals can work with these patients and address the issues that are coming into play.

7.1 Keep Food Records: Food Intake Varies from Day to Day and it is Easy to Forget to Diet Every Day

People vary in their eating patterns from day to day. Weight reduction prescriptions are such that the patient should aim for a caloric deficit of approximately 500 calories per day, which would achieve weight loss of about one pound per week. This can seem like somewhat of an abstract since the patient may not know how many calories he/she is eating in the first place. Since most people vary in their food intake from day to day, they have difficulty recognizing if they are eating less than they were previously. For this reason, simply urging patients to "eat less" of certain foods in general, is unlikely to produce clinically significant weight loss. Specific advice is more appropriate and easier for the patient to achieve. Examples of specific actions that decrease caloric intake include, cutting down portion sizes of high calorie, frequently consumed foods; avoiding appetizers; eliminating a second cocktail; replacing a second serving of steak at dinner with vegetables; or ordering roasted, baked, grilled, or steamed foods instead of fried, deep fried, sautéed, or creamed items when dining out.

Patients should also be advised to increase their intake of foods that are low in totally calories while also increasing the fiber content of their foods and replacing high-fat food with minimally processed, carbohydrates or proteins. A diet rich in food that is low in energy density, such as fruits, vegetables, and soups, will reduce caloric intake while also promoting satiety. This strategy is thought to be superior to a fat and portion restricted diet. A 2010 study evaluated the energy density of daily intake for three different groups. One group comprised overweight adults, another was of normal weight adults and the third consisted of weight loss maintainers (who had lost  10% of maximum body weight and kept it off for  5 years). Dietary intake was collected via three 24-hour phone dietary recalls and energy density was calculated using three different methods. Results showed that those in the weight loss maintainer group consumed significantly less energy per day (49). When the energy density of food is decreased, but the volume of food remains the same, calories consumed will decrease. In one study, the energy density of foods was lowered by 30%, consequently, daily energy intake also decreased by 30% (77).

Providing calorie recommendations, and instructions to keep food and physical activity records, will help patients see what factors influence their weight. Patients who record their daily food intake (i.e., food item, portion, calories, time of eating, and fat grams, if desired) as well as their physical activity for the day, are more successful in weight loss and weight maintenance efforts than those who do not (78). Some patients find it helpful to write down their emotions during their meal times to help assess whether the patient is eating out of emotion or out of hunger. The National Weight Control Registry data indicates that frequent self-monitoring of caloric intake and weight helps patients to maintain their new lower weight (79).

Patients who are able to self-monitor are more successful in weight loss efforts than those who do not self-monitor. Self-monitoring fosters awareness, an essential initial step in behavioral change. The Handbook of Assessment Methods for Eating Behaviors and Weight-Related Problems states that, “It is well established that self-monitoring or recording daily intakes via food records is a useful tool in weight loss programs” (80). Furthermore, the interplay among awareness, self-observation, recording, and self-evaluation can enhance self-management by improving how individuals attend to their health. A common denominator among all successful weight losers is self-monitoring (57;78;79;81). Patients who use food records report they have a heighted awareness of their eating behaviors, they recognize the need to make significant dietary changes and they are more able to “stay on track.” In addition, their label reading, fat and calorie counting, and portion determination skills are improved (81). These are all important skills that overweight or obese individuals need to lose weight and/or maintain their weight at lower levels. These are also skills for making healthier lifestyle choices throughout the rest of their lives, thus improving their overall health.

 

7.2 Self-reports of Energy Intake are Almost Always Underestimated

The average, healthy, adult, American male consumes approximately 2,800 calories per day, and the average female about 1,800 calories. Yet, such intakes are seldom accurately reported in a diet recall or food log. Usually, the recall or food log will show a much lower calorie intake. Reporting energy intakes is difficult, even for individuals who have been trained to do so accurately. In other words, people are widely unaware of what or how much they are eating on a daily basis. Even small omissions or inaccurate portion size reports could subtract hundreds of calories from the total calorie consumption of the day. Several days of observation are necessary to achieve accurate calorie intake since energy balance is achieved over weeks, not days. Thus, a report from any given day is certain to have a considerable amount of random as well as probably systematic error when used to estimate usual calorie intake.

Underreporting of energy intakes is common and usually off by a large margin (20%) in virtually every patient, and particularly so among the overweight population (82). Objective biomarkers of energy output such as doubly labeled water indicate that underreporting could be as great as 1,200 calories per day in very obese persons (83;84). Subjective reports of energy intake are often so low that if they were actually true, those patients should be losing weight. But in fact, they are gaining weight. It is biologically impossible to gain weight on a hypo-caloric diet, so underreporting must be considered (85).

The most common problem arising from these errors is that patients’ actual weight loss is usually less than was desired. This could be a result of underreporting and underestimating intake. Typically, when overweight people report their intakes by recall, they often underestimate their intakes by 30 to 40%. These patients are likely to make similar mistakes in underestimating their intakes on reducing diets because of difficulties in portion size judgment, forgetting, the social desirability of reporting adherence to the prescribed regimen, and other factors. This can be explained by the “flat-slope phenomenon,” which describes how individuals with a high intake of food tend to underreport their intake while those with a low intake of food tend to over report (48). For example, many people underestimate or forget that their very large food intake on weekends “counts”; or they forget to count alcohol, snacks, or something they may have had a second helping of. Thus, on a 1,200-calorie diet, actual consumption may be 2,600 calories or more on some days, and weight loss understandably slows.

Methods for assisting dieters in minimizing diet recall errors would be to use household measures or weighing scales to determine the portion size consumed more accurately, in addition to the use of food diaries. Portion-controlled liquid meal replacements, frozen low calorie entrees, and other foods that are fixed in portion sizes might also be helpful in not only portion control but also in reporting that intake because the portion size is usually labeled (see 10.3 Formulas and Meal Replacements).

Consistency in reporting intake does not necessarily mean that the reported intake is accurate, which is especially true for those who are morbidly obese. Underreporting is also especially pronounced among women, smokers, and those of low educational and socioeconomic status (83). In addition, those who underreport tend to be consistent under-reporters, and so they are difficult to detect since the records are all similar (83). In spite of these limitations, patients who keep food journals or diaries, are more successful in losing and/or maintaining weight loss than those who do not (78). Additionally, self-reports are useful for the patient and counselor alike to obtaining clues on dietary patterns that may be helpful in working through barriers to weight loss.

 

7.3 Remind Patients to Stay Active: Self-reports of Energy Output Tend to be Overestimated

Self-reports of energy output as measured by physical activity questionnaires have been validated using doubly labeled water methods. Some lengthy questionnaires used for research purposes are quite accurate at estimating physical activity (86). However, the shorter questionnaires, used clinically, are not accurate for individuals (87). As is the case with dietary recalls, physical activity questionnaires may be useful for self-monitoring, but should not be used for prescribing or assessing energy intakes or outputs exactly. Motion sensors (pedometers or accelerometers) have become popular in recent years. Accelerometers and pedometers provide objective physical activity measurements and are sensitive to walking (88). Either is a worthwhile purchase to help in self-monitoring of physical activity.

Accelerometers measure the body’s acceleration in one direction for long periods. In contrast to the pedometer, an accelerometer distinguishes between different walking speeds and intensities. Many accelerometers also record the amount of steps taken, allowing comparison with pedometers. Accelerometers have been validated as accurate forms of measurement in regards to steps counted (88). On the other hand, pedometers are less expensive because they measure only step count and not walking speed so this may be a better option for someone who is not willing or able to purchase an accelerometer. Unfortunately, pedometers are less sensitive to detecting steps if a person is walking slowly (e.g., less than two miles per hour). When walking at this slow of a pace, pedometers underestimate step count by approximately 50 to 90%. However, if a person is walking above 3.5 miles per hour, most pedometers approach 100% accuracy in step count (89). If a patient chooses to purchases a pedometer, piezoelectric pedometers are best because they are more accurate than spring-levered pedometers. This is especially true for obese or elderly individuals, who are more likely to walk at a slower pace, because these pedometers measure slow walking speeds more accurately (89;90).

If a motion sensor, either a pedometer or accelerometer, has a calorie counter built in, it could be inaccurate. Rather than focusing on the calories burned according to the motion sensor, patients should focus on the number of steps walked per day. Goals of a certain number of steps each day can be prescribed and patients are able to monitor their progress on their own. One common step count goal is 10,000 steps per day. However, most patients need to work up to that number so they should set smaller, more achievable goals until they feel they can reach the ultimate goal of 10,000 steps. The basic point is that if the patient can be induced to walk 10,000 steps a day or to gradually increase the number of steps he walks no matter how low it is, progress is being made, and the finer points of absolute accuracy can be disregarded. The ability for patients to quantify physical activity in a tangible way fosters commitment, encourages performance, provides a realistic goal, and eventually may provide a feeling of self-accomplishment.

 

7.3.1 Compensatory Decreases in Energy Output Occur on Most Reducing Diets

Unconscious compensatory decreases in physical activity usually occur on reducing diets, particularly if diets are at a very restricted calorie level. These decreases in physical activity result in slower weight loss. As a rule of thumb, if a person is decreasing calorie intake by 500 calories, there is a corresponding decrease in energy output of about 165 calories. This is a result of decreased resting metabolic rate, decreased discretionary physical activity and a decreased in energy used moving the body, which results in only at 335-calorie deficit rather than a 500-calorie deficit. So, the caloric deficit may be less than anticipated and actual weight loss becomes less than desired (91). Energy balance seems to be more strongly preserved during energy deprivation than during energy surplus, which impedes weight loss to a greater extent (92-94). This retarding effect on weight loss may be due to compensatory slowing down of resting basal metabolic rate, decreased non-obligatory physical activity and decreased thermogenesis. By including physical activity during the weight loss phase, these alterations can be alleviated to some extent, through greater energy output and preservation of lean tissue (10). Health professionals can use this information as an additional incentive for patients to maintain physical activity.

7.3.2 Physical Activity Guidelines for Americans who are Overweight

All weight loss programs should include physical activity. According to the 2008 Physical Activity Guidelines for Americans, recommendations for weight loss include engaging in 45 to 75 minutes of moderate-intensity activity per day. This can include activities such as, walking at least three miles per hour, participating in water aerobics, ballroom dancing, or gardening. Alternatively, individuals could instead participate in 22 minutes of vigorous activity per day, such as swimming, jogging, jumping rope, or hiking. Once an individual loses weight, physical activity and exercise are still important for maintaining weight loss. For weight maintenance, 60 minutes of moderate activity per day or 30 minutes of vigorous activity per day is recommended. In addition, weight resistance activities, which involve all the major muscle groups, are recommended for two or more days per week. If these recommendations cannot be achieved, it is helpful to stress to the patient that t some activity is better than none. These national recommendations should be used as goals but it may take time to reach those goals. It is a good idea to start where the patient feels comfortable and confident that they can accomplish their goals before overwhelming them with requirements of great amounts of exercise.

Dramatic decreases in weight often occur on reducing diets, particularly in the first few weeks. This is especially true for those on severely hypo-caloric regimes (with deficits of 1,000 calories per day), those on ketogenic diets, and those on very low carbohydrate diets (95;96). The type of fluid shift (loss or increase of fluid) depends on the caloric level and macronutrient composition of each diet. For example, high protein, low carbohydrate diets increase obligatory urine volume due to greater urinary loads of nitrogen, ketones, and other solutes (particularly sodium, if they are low in sodium), and will result in increased fluid losses . Increased loss of lean tissue is also associated with large fluid losses because of loss of nitrogen from tissues, such as muscle, along with water, which comprises most of the lean tissue. Fluid losses are also more apparent on hypocaloric regimes that are very low in carbohydrate (<100 grams/day and especially <50 grams of carbohydrate), since they are insufficient to replete glycogen stores, and glycogen consists largely of carbohydrate and water. Shifts in water balance may cause very dramatic deviations from the usual linear slope of weight loss. They may also cause rapid weight accumulation over just a few days if there is a period of non-adherence. This is a result of the body storing glycogen and water when the dieter eats a large carbohydrate load after a period of carbohydrate deprivation. Fluid accumulates rapidly because for every gram of glycogen that gets stored, three grams of water are stored with it. Thus, gains or losses of glycogen are associated with large changes in body water balance and water weight. These shifts can be sudden and alarming to patients (96).

7.4.1 Weight Loss Varies with Water Balance Shifts

Water balance and weight often shift during the initial period of weight loss programs, particularly on very low carbohydrate diets. As glycogen stores are depleted in response to reduced carbohydrate intake, there is an increase in fluid loss, which produces an initial and often dramatic weight loss. However, the steep rate of initial weight loss will not continue. Patients should be reminded that one-half to one pound per week of fat loss is a realistic, achievable goal that will improve their health. Additionally, they should be told that it is difficult to continue losing weight at such a rapid pace. Since glycogen stores are likely depleted, and the initial diuresis has already been achieved, there is less water weight to lose. A recent position paper on Weight Management concluded,

7.4.2 Remind Patients that Fat Loss and Weight Loss Do Not Always Track over the Short Run, Although They Do Over the Long Run

In the long term, fat loss and weight loss closely parallel each other. However, this is not necessarily true at the beginning of obesity treatment. The amount of weight that is lost over time, particularly in the beginning (the first several days) depends not only on the energy deficit from metabolic needs, but also on adherence to the weight reduction plan (76). Additionally, shifts in water balance may be considerable over the short run. These shifts can make it seem as though more fat is being lost than in reality. It is important to educate patients about these patterns so they are aware of what is happening to their body as they go through their weight loss program.

8. Set the Caloric Level of the Reducing Diet

If a caloric level of a weight-reducing diet is to be set, the two general approaches include calculated-deficit and fixed-calorie diets. With calculated deficit diets, current daily caloric needs of the patient are either measured or estimated, and the deficit is derived by subtracting from those needs (70). Fixed-calorie diets have a pre-determined recommendation for total daily caloric intake, based on caloric levels that produce weight loss in clinical trials (70) Fixed-calorie diets include low-calorie diets (LCD) and very-low-calorie diets (VLCD). Calculated-deficit and fixed-calorie approaches are discussed in this section. Another approach, in which an ad libitum diet designed to produce a caloric deficit through restriction or elimination of particular foods, is described in section 9.

8.1 Calculated-Deficit Diets

From the clinical standpoint, hypocaloric diets must be defined in terms of the energy needs of the individual and the deficit that will be created, since it is the size of the energy deficit that will determine the physiological effects expected. Ideally, energy needs should be based on resting metabolic rate (RMR), while taking into account level of physical activity. If possible, RMR should be measured (e.g., with indirect calorimetry) (70), since it can vary significantly from prediction calculations in obese individuals (97). Products such as the MedGemTM indirect calorimeter can be used in an office environment to quickly, easily, and accurately estimate a patient’s RMR (98), although caution should be used, since it may over-estimate RMR in overweight individuals [Anderson et al., 2014]. However, “if RMR cannot be measured, then the Mifflin-St. Jeor equation using actual weight is the most accurate for estimating RMR for overweight and obese individuals” (57;70). According to the Academy of Nutrition and Dietetics Evidence Analysis Library (EAL), the Mifflin-St. Jeor equation accurately predicted RMR using actual body weight within +/- 10% of measured RMR in 70% of obese individuals (57). Of the remaining 30%, 9% were overestimations and 21% were underestimations. Table 12 presents this equation. The individual error range was a maximum overestimate of 15% to a maximum underestimate of 20%” (24). While the Harris-Benedict and WHO equations are often used in clinical practice with reasonable accuracy, results have been mixed regarding their applications to individuals who are overweight or obese.

After calculating the patient’s RMR, his/her RMR should be multiplied by an appropriate physical activity factor to provide a baseline daily caloric level for weight maintenance. Once a baseline caloric level is configured, the patient’s recommended calorie intake should be reduced to facilitate weight loss. Reducing the calorie level by 500 calories is a common strategy to yield a weight loss of approximately one pound per week, although reductions of up to 750 calories per day are sometimes used (31). Another approach is to reduce current caloric intake by 30% (31). However, depending on the patient’s BMI and current intake, a larger reduction in calories may be needed, as described in the following sections. Calculations for estimating energy needs and various physical activity factors are provided in Table 12.

Table 12. Estimating Resting Metabolic Rate
Using the Mifflin-St. Jeor Equation
(15;57;70)
Males >19 years old
RMR = (9.99 X actual weight*) + (6.25 X height*) – (4.92 X age) + 5*use weight in kilograms ( kilogram), height in centimeters (cm).
Females>19 years old
RMR = (9.99 X actual weight*)+ (6.25 X height*) – (4.92 X age) – 161*use weight in kilograms (kilogram), height in centimeters (cm).
Activity Factors for Different Physical Activity Levels
  SedentaryLight physical activity associated with typical day-to-day life. Low ActiveWalking about 1.5 to 3 miles per day at 3 to 4 miles per hour, in addition to the light physical activity associated with typical day-to-day life. ActiveWalking more than 3 miles per day at 3 to 4 miles per hour, in addition to light physical activity associated with typical day-to-day life: 60 minutes of at least moderate intensity physical activity Very ActiveWalking more than 7.5 miles per day at 3 to 4 miles per hour, in addition to light physical activity associated with typical day-to-day life: 60 minutes of at least moderate to vigorous intensity physical activity
Males 1.00 1.11 1.25 1.48
Females 1.00 1.12 1.27 1.45

The major determinant of weight loss on reducing diets is size of the actual, and not the prescribed, caloric deficit. Thus, if energy needs can be measured or estimated with reasonable accuracy, then a calculated caloric deficit would be the preferred method (70). Once caloric needs for current weight maintenance are determined, the deficit can be calculated by subtracting 500-750 calories, or with a 30% caloric reduction. Diets that reduce caloric intake relative to energy expenditure result in weight loss, regardless of macronutrient composition (31).

Recent advances in methodology such as mathematical modeling have demonstrated the amount of weight loss may not be as high as one might predict based on energy balance alone (99). This is due largely to metabolic slowing that occurs with weight loss, even after accounting for declines in lean tissue (100). As weight is lost, total energy expenditure declines; so fewer calories are needed to maintain weight (101). Thus, ideally, RMR should be measured periodically as weight is lost, and caloric intake recommendations adjusted accordingly. Physical activity may ameliorate some of this decline, but not all (100).

8.2 Low-Calorie Diets (LCDs): 1,000 to 1,200 Calories for Females, 1,200 to 1,600 Calories for Males

The caloric level of the diet requires attention first and foremost; after this, other characteristics of the diet can be considered. Diets that reduce caloric intake to about 1,200 to 1,500 calories in women, and 1,500 to 1,800 calories in men will result in weight loss if they are adhered to perfectly, regardless of their macronutrient composition (102; 31). This is because such caloric levels will result in a caloric deficit for most overweight and obese adults. The National Institutes of Health have recommended low calorie diets of 1,000 to 1200 calories for women, and 1,200 to 1,600 calories per day for men, although adherence may be difficult with lower caloric intake (24;65). Either way, the rationale is that on such regimens, a deficit of approximately 500 to 1,000 calories per day will be created, which should result in a slow progressive weight loss of one to two pounds per week. Two sample menus and other materials at 1,200 and 1,600 calories and many aids to assist the physician are provided in the NIH monograph (65). The ChooseMyPlate.gov website also provides such materials and worksheets that can be accessed by consumers, and guide them daily through their weight loss.

It is important to recognize that when using fixed calorie reducing diet plans, that even with perfect adherence, individuals will vary greatly in their weight loss. This is because their resting energy needs and physical activity, and thus energy outputs, often differ markedly, and may fluctuate even within an individual.

The National Institute of Health’s Obesity Initiative sponsored an evidence based review of low calorie diets (65). It found that on average, diets such as these reduced body weight by an average of 8% over three to 12 months of treatment, and that the losses were accompanied by decreases in abdominal fat, which is the type of adipose tissue deposition that is associated with highest chronic disease risk. However, no improvements were noted in cardio-respiratory fitness as measured by VO2 max unless the dieters also increased their physical activity (24).

There are many pre-packaged meals on the market that fit into the low-calorie diet category, including Healthy Choice®, Lean Cuisine®, and Smart Ones®. Some patients find them useful for one or more meals a day since they provide a measured and moderate amount of calories per meal. Weight Watchers®TM, Jenny Craig®TM, and NutriSystemTM are more structured commercial programs that also can provide pre-packed, pre-portioned food options. See Tables 21 and 23 for a list of these programs and products.

 

8.3 Very-Low-Calorie Diets (VLCDs): ≤ 800 Calories

VLCDs supply 800 calories or less, a total of 50 to 80 grams of protein, 100% of the Reference Daily Intake (RDI) for vitamins and minerals per day, and are designed to produce very rapid weight loss while still preserving lean body mass (39). VLCDs are prescribed as a form of intensive diet therapy, which require close medical supervision, and should not be used long-term. They are intended to induce quick and significant weight loss of about 3-5 pounds weekly, or 14 to 21 kilograms over a short time (11-14 weeks). VLCD’s are sometimes used to provide a jump-start to further obesity treatment. This is typically done through meal-replacement liquid diets (6), as described below. Any diet, regardless of its caloric level, that provides less than half of an individual’s energy needs can be considered a VLCD for that individual. However, virtually all adults have energy needs that exceed 1,000 calories per day, and therefore any diet below 500 calories, and for most individuals, diets below 800 calories, are VLCDs. Depending on a person’s caloric requirements, other regimens that are higher in calories may also be VLCD for some people with very high energy needs using this same rule of thumb; for example, a 1,200 calorie diet prescribed to a man whose usual intake is 3,000 calories would also qualify as a VLCD..

8.3.1 Uses and Candidates for Therapy

These VLCDs are reserved for special uses and for individuals at high risk because of their potential for greater adverse metabolic effects and the consequent need for more extensive medical monitoring. Possible side effects range from fatigue, constipation, nausea, or diarrhea to more serious risks such as ketoacidosis and gallstones (section 8.3.4). VLCDs are often used when the health risks from obesity are particularly acute and threatening so that it is imperative to lose weight. Other individuals can usually reduce just as well on a LCD with less risk and discomfort (103). According to the National Task Force on the Prevention and Treatment of Obesity, VLCDs in patients with BMIs >30 are usually effective in promoting significant short-term weight loss, in addition to improving coexisting obesity-related conditions (e.g., obstructive sleep apnea, poorly-controlled type 2 diabetes, hypertriglyceridemia) (104). However, these diets require close metabolic monitoring (~at least every 2 weeks), and should only be prescribed and adjusted under the supervision of a physician specializing in obesity care. Medical contraindications include recent myocardial infarction, cardiac conduction disorders, history of cardiovascular disease, renal or hepatic disease, cancer, type 1 diabetes, and pregnancy. Behavioral contraindications to their use include bulimia nervosa, major depression, bipolar disorder, substance abuse, and acute psychiatric illness. The advantages of the VLCD for patients include a rapid improvement in blood pressure, blood glucose, serum lipids and often-psychological status. For those who require surgery, the rapid loss of weight may reduce some of the surgical risks associated with obesity.

 

8.3.2 Formulations Available

The hallmarks of the VLCD are the low calorie level and a relatively high percent of protein; which is at least 0.8 grams, but up to 1.5 grams per kilogram of ideal body weight (39;105). Protein needs are elevated on VLCD because in the hypocaloric state, the efficiency of protein utilization for maintaining the body’s lean cell mass is lessened since some of the amino acids are metabolized to produce glucose. Also, very heavy people who often are candidates for therapy have a larger lean body mass, and thus more lean tissue, as well as much more fat than their smaller peers. Even after adjustment for their greater fatness, total protein needs, which are most highly associated with the size of the lean body mass, are elevated. Therefore, higher protein levels may help to preserve protein nutritional status, although this remains to be demonstrated. VLCDs also have extremely low fat content and relatively low carbohydrate levels, making them ketogenic. Without special formulation or supplementation, the VLCD is deficient in several vitamins and minerals, specifically potassium, calcium, iron, zinc, vitamin C, vitamin B6, copper, and possibly other nutrients.

There are two major types of VLCDs currently in use; commercial and "home-made" preparations. The commercial preparations include powdered products that are rich in egg- or milk-based proteins, are mixed with water, and consumed four to five times per day. The commercial products must provide at least 70 grams of protein by law, and often contain much higher amounts of high quality protein (70 to 100 grams), 50 to 100 grams carbohydrate, and up to 15 grams fat per day, plus vitamins and minerals in amounts to meet the Recommended Dietary Allowances (RDA). These products are formulated under FDA regulatory specifications. They are convenient and have a predictable and adequate composition when used as directed. Their major disadvantage compared to home preparations is their higher cost. The formulas or prepackaged meals are relatively choice-free and help dieters avoid contact with conventional foods, which in some cases may facilitate dietary adherence and remove temptation.
Several commercial weight loss programs are available that provide an entire program of commercially prepared VLCDs plus the other essential aspects of a sound weight control program, including dietetic advice, exercise, behavioral modification, and supervision during the VLCD and post VLCD phases. The choices include the programs of HMRTM (Health Management Resources), OptifastTM (Novartis Nutrition), and MedifastTM. These programs employ health professionals who are trained in weight management, and a structured program that encourages adherence. The major disadvantage is that they are expensive ($3,000 to $34,000 for 26 to 28 weeks), and costs may not be covered by health insurance (105). Also, there is the uncertainty that the weight that is lost will remain so over the long run. Therefore a serious psychological as well as economic investment of effort in long-term weight management is also mandatory. See Table 13 for available program details.

 

Table 13. Medically Supervised Meal Replacement Programs
Program/Product and Company Description Is product medically supervised?
HMR TM (Health Management Resources) The HMR Decision-Free TM Clinic Weight-Loss Program: food provided includes shakes, puddings, soups, entrees, bars, and multigrain hot cereals. Yes
Medifast TM Provides special meal-plans for women, men, patients with diabetes, seniors, and teens. Six meals per day are prescribed, and foods offered include shakes, bars, soups, scrambled eggs, oatmeal, chili, puddings, and hot and cold drinks. All products are suitable for people with type 2 diabetes. Yes
Optifast TM (Novartis Nutrition) Comes as a powder to be mixed with water or as a liquid ready-to drink beverage. Patients are prescribed 5 packets of formula every 3-4 hours per day, in place of meals. No

The "home made" VLCD regimens are sometimes referred to as "protein-sparing fasts", or "protein sparing modified fasts" (PSMF). This is a misnomer since they do not “spare” protein except in contrast to a total fast. They are usually based on lean meat, fish or poultry and a few other foods plus supplements of two to three grams of potassium chloride and a multivitamin/multimineral supplement in amounts approximating the Recommended Dietary Allowances (RDA). Without such supplementation, they may be nutritionally inadequate. When patients are provided with appropriate dietetic counseling and health supervision by a physician who is experienced in the use of VLCDs and other aspects of a complete weight reduction program, these formulations are also safe and generate rapid weight loss. The extremely hypocaloric versions of VLCDs (e.g., less than or equal to 800 calories per day), which are low in carbohydrate and sodium, promote a mild ketosis that gradually leads to diuresis and rapid weight loss in the first several days on the diet.

8.3.3 Use of Very-Low-Calorie Diets (VLCDs)

Evaluation of general health and cardiac status is important prior to the institution of a VLCD. Evaluation of medication dosages and physician monitoring during the regimen are also important, since with weight loss dosing may need to be adjusted. Many practitioners begin the regimen with a two to four week low calorie diet (LCD) phase to assess the ability to comply with a restrictive regimen, and to begin the weight loss process. This is followed by a 12 to 16 week VLCD phase; the regimen is limited to this amount of time to avoid excessive loss of lean tissue. The VLCD phase is then followed by a 12 to 14 week refeeding phase of transitioning back to usual foods and gradually increasing caloric levels. The goal is to increase calories from healthful foods up to 1,200 to 1,500 calories per day, increasing caloric intake by 100 to 150 calories per day (39). This helps to avoid rapid weight changes due to refeeding with restoration of glycogen stores and shifts in water balance. The refeeding phase also provides a time for assisting the dieter to plan a maintenance diet on conventional foods and to solidify a physical activity schedule. VLCD are most effective when administered as part of a more general weight control program that includes physical activity, nutrition education, behavioral modification and attention to decreasing other risk factors. If additional weight loss is needed, it is recommended that several months elapse before another VLCD phase is instituted (106). Although lean tissue is lost on most weight reduction diets, this is a particular risk on VLCD, since greater energy restrictions are associated with more lean tissue losses (73).

8.3.4 Safety of Very-Low-Calorie Diets (VLCDs)

The VLCD induces semi-starvation, which has both benefits and risks to the patient. Occasionally, with inadequate commercial products, such as one sold in the 1970’s that consisted of hydrolyzed collagen (an incomplete protein consisting solely of the amino acid glycine) with inadequate amounts of electrolytes, vitamins and minerals, deaths occurred (107). Today, commercial products are better regulated and are nutritionally complete by law; however, the potential for misuse still exists.
Some physiological effects are inevitable on VLCDs. On VLCDs mild ketosis occurs and increases risks of dehydration, although dehydration can be avoided by ample fluid intake. Patients on VLCDs should drink at least two liters of non-caloric liquids per day (preferably water) to make up for decreased food intake and to prevent dehydration. Avoidance of caffeinated beverages is sometimes recommended, as they can further the risk of dehydration, although moderate use is not prohibited (39). Electrolyte imbalances may occur, and so may nutrient deficiencies if measures are not taken to prevent them on "home-made" VLCD, by use of appropriate supplements. Minor side effects that occur, even with appropriate physician monitoring of cardiac and general health status, include fatigue, dizziness (due to orthostatic hypotension), muscle cramps, gastrointestinal distress (constipation and/or diarrhea), and cold intolerance. The risk of cholelithiasis (gallstones) is increased, and seems to be particularly high when weight loss is very rapid (e.g., >1.5 kilograms/week). The risk of cholelithiasis can be decreased by administering ursodeoxycholic acid, including a moderate amount of fat in the diet, and limiting the amount of weight loss to 1.5 kilograms per week (105).

 

8.3.5 Effectiveness of Very-Low-Calorie Diets (VLCDs)

Because these VLCDs are so low in energy, they usually produce a greater initial weight loss than LCDs. Patients who completed a comprehensive VLCD program including lifestyle modification lost an average of 15 to 25% of initial weight within three to four months (105). However, in comparisons of VLCDs with energy levels of approximately 800 calories versus diets at lower caloric levels of 400 to 500 calories, the lower VLCDs did not necessarily result in greater weight loss, perhaps because compensatory reductions in resting energy expenditure, discretionary physical activity, and the lack of adherence on the lower calorie regimes thwarted weight loss (108).

There seems to be little difference in outcomes between commercial and properly formulated homemade VLCDs. The NIH expert panel review of existing studies found that preservation of weight loss over the long-term (e.g., >1 year) was not different on VLCD from that of LCD since most patients gained back 30 to 50% of the lost weight. Studies of VLCDs vary in their long-term results, but weight regain is common (~3.1-3.7 kg during 21-38 weeks afterwards) (31). Combining a VLCD with behavior therapy, physical activity, and active physician follow-up may help to prevent this weight regain, and lend to greater weight loss (109). As such, the long-term advantages of VLCDs in weight control are unclear. Although weight gain is common after cessation of VLCDs, individual clinicians may decide that the expense and quick initial weight loss are worth it for the patient (39).

 

8.4 Fasting and Alternate Day Fasting

Total fasting is contraindicated for weight reduction because it causes excessive breakdown of lean tissue and ketosis. Also, the compensatory decreases in resting metabolism and physical activity on total fasts are profound and counterproductive, since they lower energy output (24;5).

Short-term modified alternate-day fasting (ADF) is a relatively new dietary strategy that has not yet received enough research attention to support the effectiveness of its use. On typical ADF diets, patients consume 25% of their energy needs on the fast day, and food intake ad libitum the next day. Many of the ADF studies do not include control groups that undertake other dietary approaches to weight loss, plus the sample sizes have been small (15-64 subjects) and the durations short (8-12 weeks) (110; 111; 112; 113; 114). However, results to date show similar weight loss and improvements in metabolic factors as compared to studies using other dietary approaches to weight loss, as well as good adherence. For example, Varady et al found that ADF was a viable diet option, helping obese patients not only to lose weight, but to also decrease their risk of coronary artery disease (CAD) (110). Emerging evidence suggests that the relative contribution of fat and carbohydrate in ADF diets does not have an impact on weight and blood lipid profiles (115). An alternative fasting regimen that has become popular among some dieters is two days of fast followed by five days of usual eat. Further research is needed with larger samples, dietary control groups, and longer durations before ADF’s widespread use for weight loss purposes, however.

An alternative intermittent fasting regimen that has become popular among some dieters is two non-consecutive days of fasting and five unrestrictive days of usual eating each week. This “5:2 Diet”, developed by Dr. Michael Mosley in the United Kingdom, has spread widely throughout Europe, and now the United States. For the two fast days, men are to eat no more than 600 kcals, and women 500 kcals. Usually this consists of a very light breakfast and dinner with little or no lunch. Anecdotally, weight loss has been similar to other dietary approaches to weight loss (~2 pounds/week) for short terms (~6 weeks). However, research is lacking on this specific type of diet, so its long-term safety and efficacy has not been tested, and its appropriateness in varying populations is currently unknown

 

9. Consider the Composition of the Reducing Diet

The composition of the reducing diet is important because it may influence both the composition of the weight that is lost and nutritional status. Several published overviews of some popular diets and the basic principles that must be considered in weight control can provide more information (72;91;102;116). Over the past several decades, the potential of varying dietary composition for purposes of weight loss has been studied extensively, yet not one universally optimal diet for all patients has emerged. “A variety of dietary approaches can produce weight loss in overweight and obese individuals” (31). The challenge for practitioners is to identify which diet would be most suited for each individual patient. This may be based on previous dieting experiences, personal food preferences, lifestyle, and other factors (57).

Dietary composition on reducing diets should be geared towards decreasing risks of nutrient inadequacy and diet-related chronic diseases. Accordingly, the diet should be adequate in all nutrients, to prevent deficiencies, while following dietary guidelines for health, performance (cognitive and physical), and well-being (24;117). Consumption of vegetables, legumes, fruits, whole grains, lean sources of protein, and water should be encouraged, with emphasis on balance and moderation (33). Diets that promote extreme restriction or unusually high intakes of any macronutrient or food should be limited to a short amount of time. Recommendations for healthful composition of weight reducing diets are outlined in the 2010 Dietary Guidelines for Americans (Table 1) and discussed further in this section.

Several studies have concluded that a reduced calorie diet results in clinically meaningful weight loss regardless of what macronutrients are emphasized (31, 118; 119). For example, in a study with 811 overweight adults placed on one of four diets, the best predictor or weight loss was dietary adherence. The targeted percentages of energy derived from fat, protein, and carbohydrates in the four diets were 20, 15, and 65%; 20, 25, and 55%; 40, 15, and 45%; and 40, 25, and 35%. After six months individuals lost an average of six kilograms, 7% of their initial body weight, and after 12 months they started to regain weight. At the end of the two-year study there were no differences in the amount of weight lost amongst participants. Changes from baseline differed among the different diet groups by less than 0.5 kilograms of body weight, and less than 0.5 centimeters at the waist. However, all of the diets reduced risk factors for cardiovascular disease and diabetes at both six months and two years (120). Similarly, a systematic review of long term randomized controlled trials comparing the Atkins, Weight Watchers, Zone, and South Beach diets showed similar modest weight loss with all four approaches [Atallah et al., 2014].

According to the most recent Guidelines for Managing Overweight and Obesity in Adults (31), which is supported by a systematic evidence review by several expert panels, clinically-meaningful weight loss can be achieved through various dietary strategies. The best predictor of weight loss is adherence to a diet that produces a negative caloric balance. Thus, practitioners and patients are challenged to work together to find the option that will best help the patient to adhere. The 15 dietary strategies identified by the expert panel are listed in the table below (31). They all produced caloric deficit, but either through prescribed calorie levels, through restriction or elimination of foods or food groups, or through targeting food groups or providing foods. The expert panel concluded that the strength of the evidence was high for these studies.

High protein (30%) Zone-type (40% carbohydrate) with 5 meals dailyEuropean Association for the Study of Diabetes GuidelinesLow carbohydrate (initially <20g/day)High protein (25%), moderated carbohydrates (45%)Low fat (10-25%) vegan-styleModerate protein (12%), higher carbohydrate (58%)Low fat (~20%)High or low glycemic load mealsLow glycemic load

Table 14: Fifteen dietary strategies identified by an expert panel as having sufficient empirical evidence to recommend for weight loss (31).
Prescribed Calorie Deficit^ Caloric Deficit through Restriction/Elimination Caloric Deficit through Targeting Food Groups or Food Provision
Lacto-ovo-vegetarian High protein (30%) Zone-type (40% carbohydrate) with 5 meals daily European Association for the Study of Diabetes Guidelines
Low calorie Low carbohydrate (initially <20g/day) High protein (25%), moderated carbohydrates (45%)  
Low glycemic load Low fat (10-25%) vegan-style Moderate protein (12%), higher carbohydrate (58%)  
Low fat (<30%), high dairy & fiber Low fat (~20%) High or low glycemic load meals  
Macronutrient targeted* Low glycemic load  
Mediterranean-style    
AHA-style step 1    
^Calorie prescription was either calculated or fixed, as described in section 8
*Macronutrients have ranged 15-25% protein, 20-40% fat, and 55-65% carbohydrate in these studies

9.1 Macronutrient Distribution

As described above, the macronutrient composition of the diet does not appear to play a major role in overall weight loss; reduced-calorie diets result in clinically meaningful weight loss regardless of which macronutrients they emphasize (121; 118). There is one exception: over the short term, low carbohydrate diets are ketogenic, and may cause a greater loss of body water than body fat (at least in the first few days of the diet). Water weight is regained when the diet ceases, or when carbohydrate intake increases. In this case, glycogen stores, which hold water, are regained. Generally, when any reducing diet is maintained over the long term, if it remains hypocaloric, it will result in a loss of body fat – regardless of the distribution of macronutrients.

Although weight loss is caused by many reduced-calorie diets, the nutritional adequacy of different calorie levels and macronutrient composition for weight loss diets varies (122). The lower the reducing diet is in calories, and the more its composition differs from usual levels, the greater the risk of nutrient inadequacy. For the most part, moderate fat, balanced macronutrient reduction diets are nutritionally adequate. Very low-fat diets tend to be deficient in vitamins E, B12, calcium, iron and zinc. High fat, low carbohydrate diets are nutritionally inadequate, and require supplementation to make them nutritionally adequate in many nutrients (96;123;124). They are often low in fiber, so constipation may occur. In this case, fiber supplements and ample water intake may be recommended. Dietary supplements used on weight reduction diets should be within RDA levels and below upper safe limits.

Metabolic parameters may improve on some various popular diets including decreased blood pressure, blood lipids, blood sugar, and serum insulin, related to energy restriction and weight loss, regardless of the macronutrient composition of the diet. However, there are some differences. Moderate fat, balanced nutrient reduction diets lower low-density lipoprotein (LDL) cholesterol, normalize plasma triglycerides, and normalize ratios of HDL/total cholesterol. High fat, very low carbohydrate diets result in ketosis. Low and very low-fat diets (e.g., 15-20% of calories) reduce low-density lipoprotein (LDL) cholesterol, and after a transient rise in triglycerides, may also decrease plasma triglyceride levels. Low carbohydrate diets (e.g., <100 grams of carbohydrate) that result in weight loss may also cause a decrease in blood lipids, blood glucose and insulin levels, and blood pressure. However, these diets are often high in saturated fat, total fat, and in dietary cholesterol, and low in plant-based nutrients such as fiber, so variability in metabolic responses may be seen, due in part to genetic predisposition. Moreover, these diets are ketogenic, often causing signs and symptoms such as diuresis, dizziness, halitosis, fatigue, weakness, hypotension, and malaise.

Hunger may vary on the different diets, and also from one individual to the next, but little objective evidence is available for comparing different reducing diets on their anti-hunger effects. Many factors affect hunger, appetite and subsequent food intake, including interactions between physiological and non-physiological factors. Schoeller and Buchholz speculate that a greater consumption of protein may increase satiety, which in turn results in better adherence to hypocaloric diets, however, substantial long-term evidence to support this supposition is lacking (124), and more research is needed in this area.

Long-term dietary adherence is likely to be a function primarily of psychological and lifestyle issues rather than macronutrient composition itself. At present little is known about the nutritional or other characteristics of diets that maximize adherence. It is likely that "one size does not fit all" in this respect, so the importance of individualization is underscored.

 

9.2 Protein

This section outlines dietary protein needs during weight reduction. The Recommended Dietary Allowance for protein is 0.8 grams per kilogram per day, but most Americans eat about 1.2 grams per kilogram per day, or approximately 15% of their total caloric intakes from protein. For people in energy balance and at a stable weight, the World Health Organization (WHO) recommends that dietary protein should account for approximately 10 to 15% of energy intake (125).

9.2.1 Protein Needs During Weight Reduction

Protein requirements tend to rise on hypocaloric diets, especially on VLCDs when protein is burned for energy. Thus, protein needs increase so that loss of lean body mass can be minimized. This is because when energy intakes are insufficient, glucogenic amino acids are used to maintain blood glucose levels and other ketogenic amino acids must be used for energy, so overall protein requirements increase. Fortunately, the hormonal milieu in hypocaloric states spares nitrogen to some extent and causes preferential use of fat for energy. However, fatty acids cannot be converted to blood glucose, so glucogenic amino acids are needed for this. Inevitably, as adipose tissue is mobilized some lean tissue is lost and consequently some nitrogen is also lost. Losses of water, calcium, phosphorus, potassium, and vitamins follow the loss of lean tissue. Excess losses of lean body mass can be hazardous, affecting cardiovascular function, exercise tolerance, and possibly immune responses, and thus should be avoided. As mentioned previously, excess loss of lean tissue can result from energy deficits that are too great (73).

As a rule of thumb, a minimum of 65 to 70 grams of protein is needed daily. On a VLCD, 1.5 grams of high quality protein per kilogram of ideal body weight per day is desirable, with intakes no less than, and preferably more than, 65 to 70 grams daily. Intakes may need to be even higher if the dieter suffers from certain diseases or is physically stressed, since nitrogen losses may be more extreme in these states. On diets providing 600 to 1,200 calories per day, daily protein intake should be at least one gram per kilogram ideal body weight per day. Reducing diets over 1,200 calories per day should supply at least 0.8 grams per kilogram ideal body weight, and more if the individual is physically active. Levels should remain this high after weight loss has stopped and maintenance has begun.

 

9.2.2 High Protein Weight Loss Diets

High protein reducing diets are those that provide more than 1.6 grams per kilogram of desirable weight per day. Self-prescribed high protein reducing diets vary in their composition from about 28 to 65% of energy, providing 71 to 163 grams of protein per day. They are currently popular as a new strategy for losing weight, and are usually quite low in their carbohydrate content. Some are clearly ketogenic, and severely limit carbohydrates to below 50 grams per day. Examples include the Doctor’s Quick Weight Loss Diet (126) The Dukan Diet (126), Dr. Atkins™’ Diet Revolution (127), The 17 Day Diet (231), and various iterations of the Paleo Diet, which is discussed in more detail below. Diets that are extremely high in protein should not be undertaken for long periods of time, since their long-term safety has not been sufficiently examined.

Other diets are extremely high in protein, very low in carbohydrate and ketogenic, but also very high in fat, such as Protein Power (128). Two other high protein diets with enough carbohydrate so that they are not likely to be ketogenic are The Zone (129) and Sugar Busters (130).

Many high protein diets include elaborate instructions that prescribe strict, structured eating schedules, and involve limited food variety and dietary flexibility. The high protein diets that are ketogenic also induce quick initial weight loss because of their low caloric level, and their diuretic effect owing to glycogen depletion, and sodium and water loss. They may also be associated with decreased appetite due to the high protein intake, since protein may show to be particularly satiating (131;132). Ketosis has long been said to reduce appetite, although little data supports this. Nonetheless, for some patients these constraints may help them to achieve and maintain low calorie intakes over the short run.

Popular high protein reducing regimens are not risk-free, however. Many of these diets advocate very high intakes of protein from meat and other foods that are also often high in saturated fat, cholesterol and sodium while they are low in dietary fiber, antioxidants, potassium, calcium, magnesium, and some vitamins. The purine content of meat, poultry, seafood, eggs, seeds, and nuts is high, and can increases uric acid levels and risk of gout in susceptible persons. The high protein load may also increase urinary calcium loss if it is not buffered (133). In patients with diabetic nephropathy, very high protein diets may speed progression, although the data are not definitive (134). Because many high protein diets are often by default low in carbohydrate, they also can cause an increase in ketosis. Finally, these diets do not necessarily promote greater long-term weight loss as compared to other options (135;136; 118).

 

9.3 Fat

This section outlines dietary fat needs during weight reduction.

9.3.1 Fat Needs During Weight Reduction

Even on reducing diets, the human body needs small amounts (e.g., three to six grams) of essential fatty acids (linoleic or arachidonic acid). Some fat is also necessary as a carrier for the fat-soluble vitamins A, D, E, and K. Therefore the diet should not be devoid of fat. However, because fat is calorically dense, it is often decreased on reducing diets to reduce energy intake while increasing bulk.

9.3.2 Moderate to Low-Fat Balanced Deficit Reducing Diets

In general, levels of dietary fat, saturated fat, trans fat, polyunsaturated fat, monounsaturated fat, and cholesterol should follow guidelines from the American Heart Association (AHA) on weight reduction diets. While lower levels may be appropriate in some cases, they amply meet requirements while supporting cardiovascular health (24).
Weight reduction diets that are moderate to low in fat (20 to 30% of calories) are called "balanced deficit" diets because they maintain a reasonable balance among macronutrients similar to that recommended in MyPlate, DASH, and the Dietary Guidelines for Americans (15; 117). They tend to achieve most of the caloric deficit by reducing fat from the typical level in North American Diets of about 34% or more of calories to 20 to 30% fat, 15% protein, and 55 to 65% of calories from carbohydrates. Some examples of balanced deficit diets are the Weight Watchers® Diet (25% fat, 20% protein, and 55% carbohydrate, with 26 grams of dietary fiber), Jenny Craig®, the National Cholesterol Education Program Step 1 diet (25% fat), diets based on the MyPlate, the DASH diet, the Shape up and Drop 10 diet of Shape Up! America (33), and the Nutrisystem® diet. Popular diet books using this approach include The Biggest Loser Diet (232), The Mayo Clinic Diet (233), and The Engine 2 Die (234). These dietary patterns have been extensively reviewed and appear to be effective for weight reduction on low calorie diets for most individuals.

 

9.3.3 Very Low-Fat Reducing Diets (<20% Fat Calories)

Very low-fat diets such as the Pritikin Diet (137), the Ornish Diet (138), and The Spark Solution Diet (235) have been advocated not only for weight reduction, but also for improving cardiovascular risk profiles. The Ornish Diet, which is very low in fat (13% of calories) and saturated fat, very high in carbohydrate (81% of calories) and very high in fiber (38 grams), is part of a program that includes nonsmoking, exercise and behavior modification. It was shown to reduce some cardiovascular risk factors in a limited long term study (138). For those who can adhere to the Ornish regime it may be helpful. However, it may not be appropriate for all populations, such as diabetics.

9.3.4 High Fat Diets for Weight Reduction (55 to 65% Fat)

High fat reducing diets are also usually low or very low in carbohydrate (<200 grams carbohydrate per day). Some current examples include Dr. Atkins™’ Diet Revolution (127), Protein Power (128), the Carbohydrate Addicts Diet (139), Dr. Bernstein’s Diabetes Solution (140), Life Without Bread (141), the Pennington Diet (141), and the Bulletproof Diet (236). There is some evidence that free-living, overweight people who self-select high fat, low carbohydrate diets consume fewer calories and lose weight (102). This is not because the laws of thermodynamics are violated, but because they have far fewer food options, if they adhere to such rigorous regimens. When high fat, low carbohydrate reducing diets are fed they also tend to cause ketosis and diuresis. They may also result in decreased blood lipids, glucose and insulin, along with and decreased blood pressure, but only if weight is lost. Over the short term (a few days or weeks) high fat, low carbohydrate, ketogenic diets cause a greater loss of body water than body fat, but water balance is quickly restored when carbohydrate intakes increase or when the diet ends. High fat, low carbohydrate diets are often nutritionally inadequate, so they often require some supplementation with micronutrients and fiber. If such high fat levels are continued on a chronic basis after weight is lost, they are may increase dietary risks for coronary artery disease. More research is needed in this area.

9.4 Carbohydrates and the Glycemic Index

The following section reviews carbohydrate needs during weight reduction, and the glycemic index.

9.4.1 Carbohydrate Needs in Weight Reduction

Carbohydrate needs for most individuals are at least 50 grams per day, to fuel the central nervous system, red blood cells, and other glucose-dependent tissues. If carbohydrate intake fall below this, than gluconeogenesis is likely to ensue. The carbon source for gluconeogenesis cannot be fatty acids, so in these cases, amino acids are used to maintain blood glucose and fuel glucose-dependent tissues. At least 100 grams carbohydrate, and preferably carbohydrate within the Acceptable Macronutrient Distribution Ranges (AMDR) of 45 to 65% of total energy intake, should be provided for diets that are over 800 calories per day. Under experimental conditions, both hypocaloric diets very high in sugars (mono-and di-saccharides) and diets very high in starches (digestible polysaccharides) that are equi-caloric have similar weight loss effects (143;144). However, from the practical standpoint, since many products that are high in sugar are calorically dense and often are also high in calories, added fat, and low in fiber, vitamins and minerals; sugars are usually limited on reducing diets. Additionally, high sugar diets may increase some cardiovascular risk factors (145). From the physiological standpoint, “added” sugar, and sugar inherently in the foods, are similar in their caloric contributions.
.
Individuals assigned to a low-carbohydrate, high-protein diets do lose more weight at six months than those on low-fat, reduced-energy diet. However, this difference is no longer significant at 12 months (70;116;120). An evidence review from the Academy of Nutrition and Dietetics concluded that, “An individualized, reduced calorie diet is the basis of the dietary component of a comprehensive weight management program. Reducing dietary fat and/or carbohydrates is a practical way to create a caloric deficit of 500 to 1,000 kcal below estimated energy needs and should result in a weight loss of one to two pounds per week” (70). Whether an individual dieter reduces fat or carbohydrate does not matter. If calories are similar, in the long run, weight loss amounts to reducing caloric intake. Concerns regarding an increase in cardiovascular risks with low-carbohydrate diets now do not appear to be as problematic as first thought (70). If an obese person loses weight, cardiovascular risk factors usually improve (31).

 

9.4.2 Low Carbohydrate Diets (<100 grams Carbohydrate)

Diets providing less than 100 grams of carbohydrate per day, and especially those with less than 50 grams carbohydrates per day, are ketogenic. Ketosis can be a problem on some popular diets that are very low in carbohydrates, such as Dr. Atkins™’ Diet Revolution (127), Protein Power (128), The Dukan Diet (126), Dr. Bernstein’s Diabetes Solution (140), The Bulletproof Diet (236),, and the Pennington Diet (142). Also, VLCDs containing fewer than 100 grams of carbohydrate per day are ketogenic and may lead to excessive protein breakdown to maintain blood glucose levels unless protein intakes are increased. When the body must rely on catabolism of protein to preserve blood glucose levels via gluconeogenesis, the catabolism of the protein is accompanied by loss of water. For every gram of protein (or glycogen) that is broken down, three grams of water are released, causing rapid weight loss but also a state of relative dehydration (95). Relative dehydration caused by ketosis and failure to drink adequate amounts of fluids is not only undesirable for health reasons, it reduces exercise tolerance (67). It also does not address the primary purpose of the weight-reducing strategy, which is to decrease excess adipose tissue and not water weight. One main concern with studies to date on low carbohydrate diets is that most of them to not include exercise. Since exercise tolerance declines with low carbohydrate intakes and reduced glycogen stores, more research is needed in this area before such diets can be recommended.

9.4.3 Low Glycemic Index Diets

The Glycemic Index (GI) was originally developed for the therapy of diabetes, but it has gained popularity in weight management. The GI describes the blood glucose response resulting from consumption of a defined amount of carbohydrate (usually 50 grams) from a given food, relative to the response of the same amount of carbohydrate from a control food (usually white bread)(146). In brief, the GI is an alternative system for classifying carbohydrate-containing foods according to their postprandial blood glucose responses to portions containing standardized amounts of carbohydrates (30). Since the GI is based on standardized portions, glycemic load (GL), the product of GI and carbohydrate amount, is used to evaluate the effect of meals/snacks—differing in both quality and quantity of carbohydrates—on postprandial glycemia (30).

The basic premise is that more moderate blood glucose and metabolic responses from low-GI foods and a low GL will sustain satiety and energy balance to a greater extent than would high-GI foods and a high GL load. The GI may be important in regulating hunger, voluntary energy intake, and satiety. A high-GI meal or snack may compromise glucose uptake following a subsequent meal—a phenomenon known as the “second-meal effect.” The underlying mechanism likely involves decreased insulin sensitivity with increased concentrations of circulating free fatty acids during the late postprandial phase. With regard to a high-GI meal or snack, it is thought that, “the drop in blood glucose during the middle postprandial phase may increase the preference for high-GI foods, leading to repeated cycles of excess hunger followed by hyperphagia that may last for several hours following restoration of euglycemia. These vicious cycles, exacerbated by the second-meal effect may contribute to disappointing long-term weight control with conventional low-fat diet prescriptions that emphasize the importance of consuming starchy foods” (30). Hence, low-GI foods are thought to help minimize blood glucose fluctuations, hunger hormones, and increase satiety.

Some, but not all, studies have conducted a follow-up period at 12 months showing that overweight or obese individuals on low-GI diets lose slightly more weight (1-3 kg) than those on high-GI diets or conventional energy restricted weight loss diets (147). Beyond short term weight loss, low-GI diets have also shown to decrease fasting glucose and insulin levels, reduce circulating triglycerides, and improve blood pressure (30;148;149). Thus, low-GI and low-GL diet plans may help some individuals lose weight, typically one to two BMI units, and help improve metabolic parameters and risk of cardiovascular disease. (148). The effects of low GI carbohydrates may also help to prevent excess weight gain, although more research must be done on their longer-term efficacy (150-152).

Consumption of whole grains, legumes, fruits, vegetables, and whole foods that are low in GI, is helpful in meeting fiber goals and may be helpful in weight management. A well balanced, hypocaloric low glycemic index diet may prove to be effective in properly educated, adherent patients who are willing to take the time to learn about high- and low-glycemic foods, and who do not completely exclude healthful high glycemic foods. For example, sausages, ice cream, and chocolate cake with frosting are all low GI foods, while parsnips, carrots, bananas, dates and potatoes tend to be high GI foods. This underscores the point that more than just GI must be considered in food choices and patients need to be educated accordingly. Since GI is not listed on food labels in most countries, and since many factors influence it, such as cooking, ripeness, and the other foods consumed at the same meal, this dietary approach may pose a challenge for some patients. Any reducing diet must be viewed as a whole. In the USA nutrient fact labels are available on most processed foods, and provide information on carbohydrate content. Therefore the GI does not offer much additional information. However labeling of carbohydrate subtypes is not done in some countries, and use of the GI is popular. As seen in the table at the beginning of this section, diets with altered GI have been applied in cases where calories were prescribed, where food was provided, and where intake was limited, and all produced similar results to other dietary approaches. However, data are limited in US populations. In summary, diets based on the GI may offer some patients benefits in terms of short and long-term weight loss, but the perpetuity of the regimen remains in question.

 

9.4.4 Paleo Diet

The Paleo Diet has gained considerable popularity among consumers in recent years, although no large-scale scientific study has thoroughly investigated it yet, especially for the purposes of weight loss. This dietary approach suggests that individuals consume only foods and beverages that presumably made up the diets of Paleolithic humans (153). Thus, all processed foods and beverages are eliminated in addition to grains, dairy, and legumes. Allowed foods include meats, fish, poultry, vegetables, fruits, and nuts (not peanuts). Deficits in caloric intake are achieved through elimination of large amounts of usually-consumed food types. Advantages are that this diet tends to be high in nutrients that come from vegetables and fruits (e.g. vitamin C), and it is low in sodium and glycemic index / load (154). However, it is low in calcium, and high in cholesterol (154). While it provides satiety, many subjects find adherence difficult, due to the restriction of so many foods and beverages. To date, most studies have only tested this diet short term (10 days to 3 months), with low subject numbers (9-20), sometimes lacking control groups, and without a concurrent exercise prescription (155; 154; 156; 157). Weight loss has been comparable to other similar dietary approaches (156), and improvements have been noted for several risk factors of cardiovascular disease and type 2 diabetes (154; 157), even without weight loss (155). Patients attempting this diet while on warfarin treatment should consult their physician or dietitian due to the high vitamin K coming from the abundance of vegetables (153). More work is needed to determine long-term outcomes and adherence for this diet. Particular attention should be given to bone and gastrointestinal health, since dairy, cereal fibers, and legumes are missing from this diet [158; 159; 160).

9.4.5 High-Fructose Corn Syrup and Weight

High-fructose corn syrup (HFCS) does not contribute to overweight or obesity any differently than do other energy sources (161). HFCS has been blamed for the obesity epidemic mainly due to the association between American’s increases in weight along with the increase in HFCS in our food supply since the 1970s, but similar associations are also present with bottled water sales. However, association is not causation. In 2004, Bray et al (162) hypothesized that HFCS was a direct causative factor for obesity. However, to date, there is no scientific evidence supporting this theory. As stated in White’s article from The American Journal of Clinical Nutrition, “The HFCS-obesity hypothesis of Bray et al relies heavily on the positive association between increasing HFCS use and obesity rates in the United States. However, Bray et al treated this association in isolation, offering no perspective on trends in total caloric intake or added sweeteners use in comparison with use of other dietary macronutrients.”

HFCS was introduced to the food industry in the late 1960s and was well received because it is stable in acidic foods and beverages, is easily transportable and is sweet. HFCS can be pumped from delivery vehicles to storage and mixing tanks, requiring only simple dilution before use. Furthermore, it has remained relatively inexpensive. Its sweetness mirrored that of sugar. Contrary to popular belief, HFCS is not sweeter than sucrose. The forms of HFCS in the food supply are HFCS-55 and HFCS-42 with 55% fructose and 42% glucose, and 42% fructose and 53% glucose, respectively. The remaining carbohydrates are free glucose, maltose, and maltotriose. A similar ratio of fructose to glucose as in HFCS is also in honey, invert sugar, fruit, and fruit juices (163). Table sugar or sucrose is composed of 50% fructose and 50% glucose. Hence, the ratio of glucose to fructose in both HFCS and sucrose is essentially 1 to 1. Furthermore, HFCS and sucrose both contain four calories per gram. Existing theoretical and empirical evidence suggests that fructose-induced problems are not more related to HFCS than sucrose intake (164). Total caloric intake is positively associated with BMI, independent of sugar intake (165). However, the World Health Organization and U.S. Dietary Guidelines recommend avoidance of excess added sugars due to their ‘empty calories’, which may dilute dietary quality, and also may elevate some cardiometabolic risk factors (117; 145),
HFCS is not a direct cause of the obesity epidemic in the United States. To date, there is no evidence linking these two factors (161;166;167). As Forshee et al concluded, “Evidence from ecological studies linking HFCS consumption with rising BMI rates is unreliable. Evidence from epidemiologic studies and randomized controlled trials is inconclusive. Based on the currently available evidence, an expert panel concluded that HFCS does not appear to contribute to overweight and obesity any differently than do other energy sources” (161).

 

9.5 Water

This section discusses water and electrolyte needs during weight reduction.

9.5.1 Water Needs on Reducing Diets Vary

Ample fluid intake is important on weight reduction diets to prevent dehydration, especially if diets are ketogenic, very low in calories, or being undertaken in hot climates or with physical exertion. As mentioned earlier, losses of body glycogen and protein are accompanied by losses of body water. Intake of low-calorie or calorie-free fluids such as water should be emphasized (168). Water needs go up with increases in physical activity, not only due to sweat losses, but also due to increased water losses due to respiration (67). The fatigue that some dieters associate with hypocaloric diets is often due in part to dehydration, especially if they have also increased their physical activity and exercise regimes dramatically. Body water losses of as little as 2% have been associated with decreased physical and mental performance, and impaired thermoregulation (168). Some dieters may be in a state of mild dehydration much of the time, and this is unnecessary and may detract from quality of life. General water recommendations average approximately 2.7 liters (91 ounces) per day for women and 3.7 liters (125 ounces) per day for men. This includes total water intake from all beverages and water in foods. It has been estimated that 20% of total water consumption comes from solid foods (169). A fluid intake plan should be incorporated in every weight loss regimen. Non-caloric sources of fluid should be emphasized; pure water and seltzers tend to be great choices.

9.6 Electrolytes

Under normal circumstances on a well-balanced diet that is not overly restrictive with energy, electrolyte balance is maintained. If an individual may be losing excess electrolytes due to high sweat or urine losses, electrolytes can usually be replaced with normal foods (67). The American diet is overly abundant in sodium. Potassium is not so abundant but can be obtained in fruits and vegetables. Examples of foods that are high in both sodium and potassium include tomato sauces and vegetable soups.
Electrolyte levels are of particular concern on VLCD, since occasionally cardiac arrhythmias have resulted from hypokalemia on such regimens (105). Since hypokalemia can be fatal, electrolyte levels must always be monitored on VLCD.

 

9.7 Vitamins and Minerals

The next section outlines vitamin and mineral needs during weight reduction.

9.7.1 Vitamin and Mineral Needs During Weight Reduction

Vitamin and mineral nutrition is critical during weight reduction and maintenance. The amounts of nutrients specified in Recommended Dietary Allowance (RDA) for an individual’s age and sex must continue to be met, even on reducing diets for all other nutrients (See Table 15 and Table 16). The lower the diet is in calories, the more likely it is that essential vitamins, minerals and electrolytes such as potassium, copper, magnesium, Vitamin E, Vitamin B6, folic acid, iron, and calcium are likely to be low. As a rule of thumb, diets below 1,200 calories per day are likely to require vitamin and mineral supplements in amounts approximating the Recommended Dietary Allowances (15). Above 1,200 calories per day, women in reproductive age groups may still need iron, calcium, and folic acid supplements, since their needs for these nutrients are high, but most other nutrient needs can be met by a well-balanced diet that follows the Dietary Guidelines for Americans (15). For this reason, foods with high micronutrient density, but low energy density are especially important to include on a reducing diet. They include fruits, vegetables, legumes, and lightly processed whole grains. Table 15, Table 16, Table 17, and Table 18 present the current DRIs for vitamins, minerals and tolerable upper levels (UL) for these same nutrients.

Table 15.
Dietary Reference Intakes (DRIs): Recommended Intakes for Individuals, Vitamins (292)

Food and Nutrition Board, Institute of Medicine, The National Academies
Life Stage
Group
Vitamin A Vitamin C Vitamin D ± Vitamin E Vitamin K Thiamin Riboflavin Niacin VitaminB6 Folate Vitamin B12 Pantothenic Acid Biotin Choline
(µg/d) a (mg/d) (µg/d) b,c (mg/d) d (µg/d) (mg/d) (mg/d) (mg/d) e (mg/d) (µg/d) f (µg/d) Acid (mg/d) (µg/d) (mg/d) g
Infants
0–6 mo 400* 40* 5* 4* 2.0* 0.2* 0.3* 2* 0.1* 65* 0.4* 1.7* 5* 125*
7–12mo 500* 50* 5* 5* 2.5* 0.3* 0.4* 4* 0.3* 80* 0.5* 1.8* 6* 150*
Children
1–3 y 300 15 5* 6 30* 0.5 0.5 6 0.5 150 0.9 2* 8* 200*
4–8 y 400 25 5* 7 55* 0.6 0.6 8 0.6 200 1.2 3* 12* 250*
Males
9–13 y 600 45 5* 11 60* 0.9 0.9 12 1.0 300 1.8 4* 20* 375*
14–18 y 900 75 5* 15 75* 1.2 1.3 16 1.3 400 2.4 5* 25* 550*
19–30 y 900 90 5* 15 120* 1.2 1.3 16 1.3 400 2.4 5* 30* 550*
31–50 y 900 90 5* 15 120* 1.2 1.3 16 1.3 400 2.4 5* 30* 550*
51–70 y 900 90 10* 15 120* 1.2 1.3 16 1.7 400 2.4 h 5* 30* 550*
> 70 y 900 90 15* 15 120* 1.2 1.3 16 1.7 400 2.4 h 5* 30* 550*
Females
9–13 y 600 45 5* 11 60* 0.9 0.9 12 1.0 300 1.8 4* 20* 375*
14–18 y 700 65 5* 15 75* 1.0 1.0 14 1.2 400 i 2.4 5* 25* 400*
19–30 y 700 75 5* 15 90* 1.1 1.1 14 1.3 400 i 2.4 5* 30* 425*
31–50 y 700 75 5* 15 90* 1.1 1.1 14 1.3 400 i 2.4 5* 30* 425*
51–70 y 700 75 10* 15 90* 1.1 1.1 14 1.5 400 2.4 h 5* 30* 425*
> 70 y 700 75 15* 15 90* 1.1 1.1 14 1.5 400 2.4 h 5* 30* 425*
Pregnancy
≤ 18 y 750 80 5* 15 75* 1.4 1.4 18 1.9 600 j 2.6 6* 30* 450*
19–30 y 770 85 5* 15 90* 1.4 1.4 18 1.9 600 j 2.6 6* 30* 450*
31–50 y 770 85 5* 15 90* 1.4 1.4 18 1.9 600 j 2.6 6* 30* 450*
Lactation
≤ 18 y 1,200 115 5* 19 75* 1.4 1.6 17 2.0 500 2.8 7* 35* 550*
19–30 y 1,300 120 5* 19 90* 1.4 1.6 17 2.0 500 2.8 7* 35* 550*
31–50 y 1,300 120 5* 19 90* 1.4 1.6 17 2.0 500 2.8 7* 35* 550*
NOTE: This table (taken from the DRI reports, see www.nap.edu) presents Recommended Dietary Allowances (RDAs) in bold type and Adequate Intakes (AIs) in ordinary type followed by an asterisk (*). RDAs and AIs may both be used as goals for individual intake. RDAs are set to meet the needs of almost all (97 to 98 percent) individuals in a group. For healthy breastfed infants, the AI is the mean intake. The AI for other life stage and gender groups is believed to cover needs of all individuals in the group, but lack of data or uncertainty in the data prevent being able to specify with confidence the percentage of individuals covered by this intake.a As retinol activity equivalents (RAEs). 1 RAE = 1 µg retinol, 12 µg ß-carotene, 24 µg ∂-carotene, or 24 µg ß-cryptoxanthin. To calculate RAEs from REs of provitamin A carotenoids in foods, divide the REs by 2. For preformed vitamin A in foods or supplements and for provitamin A carotenoids in supplements, 1 RE = 1 RAE.b calciferol. 1 µg calciferol = 40 IU vitamin D.c In the absence of adequate exposure to sunlight.d As ∂-tocopherol. ∂-Tocopherol includes RRR-- tocopherol, the only form of ∂-tocopherol that occurs naturally in foods, and the 2R -stereoisomeric forms of ∂-tocopherol ( RRR -, RSR -, RRS -, and RSS -∂-tocopherol) that occur in fortified foods and supplements. It does not include the 2S -stereoisomeric forms of ∂-tocopherol ( SRR -, SSR -, SRS -, and SSS -∂-tocopherol), also found in fortified foods and supplements.e As niacin equivalents (NE). 1 mg of niacin = 60 mg of tryptophan; 0–6 months = preformed niacin (not NE).f As dietary folate equivalents (DFE). 1 DFE = 1 µg food folate = 0.6 µg of folic acid from fortified food or as a supplement consumed with food = 0.5 µg of a supplement taken on an empty stomach.g Although AIs have been set for choline, there are few data to assess whether a dietary supply of choline is needed at all stages of the life cycle, and it may be that the choline requirement can be met by endogenous synthesis at some of these stages.h Because 10 to 30 percent of older people may malabsorb food-bound B 12 , it is advisable for those older than 50 years to meet their RDA mainly by consuming foods fortified with B 12 or a supplement containing B 12 .± In 2008, the American Academy of Pediatrics adjusted their 2003 recommendations for vitamin D in children from 5 µg per day (200 IU), beginning in the first two months of life, to 10 µg per day (400 IU) within the first few days of life. This increased recommendation is based on the amount of vitamin D that can be given safely per day to prevent or treat rickets and possibly provide additional health benefits. The 2004 DRIs have not yet been updated to reflect this.i In view of evidence linking inadequate folate intake with neural tube defects in the fetus, it is recommended that all women capable of becoming pregnant consume 400 µg from supplements or fortified foods in addition to intake of food folate from a varied diet.j It is assumed that women will continue consuming 400 µg from supplements or fortified food until their pregnancy is confirmed and they enter prenatal care, which ordinarily occurs after the end of the periconceptional period—the critical time for formation of the neural tube.Copyright 2004 by the National Academy of Sciences. All rights reserved. 2/15/01
Table 16. Dietary Reference Intakes (DRIs): Recommended Intakes for Individuals, Elements (130)Food and Nutrition Board, Institute of Medicine, National Academies
Life StageGroup Calcium Chromium Copper Fluoride Iodine Iron Magnesium Manganese Molybdenum Phosphorus Selenium Zinc
(mg/d) (µg/d) (µg/d) (mg/d) (µg/d) (mg/d) (mg/d) (mg/d) (µg/d) (mg/d) (µg/d) (mg/d)
Infants
0–6 mo 210* 0.2* 200* 0.01* 110* 0.27* 30* 0.003* 2* 100* 15* 2*
7–12 mo 270* 5.5* 220* 0.5* 130* 11 75* 0.6* 3* 275* 20* 3
Children
1–3 y 500* 11* 340 0.7* 90 7 80 1.2* 17 460 20 3
4–8 y 800* 15* 440 1* 90 10 130 1.5* 22 500 30 5
Males
9–13 y 1,300* 25* 700 2* 120 8 240 1.9* 34 1,250 40 8
14–18 y 1,300* 35* 890 3 * 150 11 410 2.2* 43 1,250 55 11
19–30 y 1,000* 35* 900 4* 150 8 400 2.3* 45 700 55 11
31–50 y 1,000* 35* 900 4* 150 8 420 2.3* 45 700 55 11
51–70 y 1,200* 30* 900 4* 150 8 420 2.3* 45 700 55 11
> 70 y 1,200* 30* 900 4* 150 8 420 2.3* 45 700 55 11
Females
9–13 y 1,300* 21* 700 2* 120 8 240 1.6* 34 1,250 40 8
14–18 y 1,300* 24* 890 3* 150 15 360 1.6* 43 1,250 55 9
19–30 y 1,000* 25* 900 3* 150 18 310 1.8* 45 700 55 8
31–50 y 1,000* 25* 900 3* 150 18 320 1.8* 45 700 55 8
51–70 y 1,200* 20* 900 3* 150 8 320 1.8* 45 700 55 8
> 70 y 1,200* 20* 900 3* 150 8 320 1.8* 45 700 55 8
Pregnancy
≤ 18 y 1,300* 29* 1,000 3* 220 27 400 2.0* 50 1,250 60 12
19–30 y 1,000* 30* 1,000 3* 220 27 350 2.0* 50 700 60 11
31–50 y 1,000* 30* 1,000 3* 220 27 360 2.0* 50 700 60 11
Lactation
≤ 18 y 1,300* 44* 1,300 3* 290 10 360 2.6* 50 1,250 70 13
19–30 y 1,000* 45* 1,300 3* 290 9 310 2.6* 50 700 70 12
31–50 y 1,000* 45* 1,300 3* 290 9 320 2.6* 50 700 70 12
NOTE: This table presents Recommended Dietary Allowances (RDAs) in bold type and Adequate Intakes (AIs) in ordinary type followed by an asterisk (*). RDAs and AIs may both be used as goals for individual intake. RDAs are set to meet the needs of almost all (97 to 98 percent) individuals in a group. For healthy breastfed infants, the AI is the mean intake. The AI for other life stage and gender groups is believed to cover needs of all individuals in the group, but lack of data or uncertainty in the data prevent being able to specify with confidence the percentage of individuals covered by this intake.SOURCES : Dietary Reference Intakes for Calcium, Phosphorous, Magnesium, Vitamin D, and Fluoride (1997); Dietary Reference Intakes for Thiamin, Riboflavin, Niacin, Vitamin B 6 , Folate, Vitamin B 12 , Pantothenic Acid, Biotin, and Choline (1998); Dietary Reference Intakes for Vitamin C, Vitamin E, Selenium, and Carotenoids (2000); and Dietary Reference Intakes for Vitamin A, Vitamin K, Arsenic, Boron, Chromium, Copper, Iodine, Iron, Manganese, Molybdenum, Nickel, Silicon, Vanadium, and Zinc (2001). These reports may be accessed via www.nap.edu.Copyright 2001 by the National Academy of Sciences. All rights reserved. 2/15/01
Table 17. Dietary Reference Intakes (DRIs): Tolerable Upper Intake Levels (UL a ) for Vitamins (130)Food and Nutrition Board, Institute of Medicine, National Academies
Life Stage Group Vitamin A Vitamin C Vitamin D Vitamin E Vitamin K Thiamin Riboflavin Niacin Vitamin B 6 Folate Vitamin B 12 Pantothenic Acid Biotin Choline Carotenoids e
(µg/d) b (mg/d) (µg/d) (mg/d) c,d --- --- --- (mg/d) d (mg/d) (µg/d) d --- --- --- (g/d) ---
Infants
0-6 mo 600 ND f 25 ND ND ND ND ND ND ND ND ND ND ND ND
7-12 mo 600 ND 25 ND ND ND ND ND ND ND ND ND ND ND ND
Children
1-3 y 600 400 50 200 ND ND ND 10 30 300 ND ND ND 1.0 ND
4-8 y 900 650 50 300 ND ND ND 15 40 400 ND ND ND 1.0 ND
Males, Females
9-13 y 1,700 1,200 50 600 ND ND ND 20 60 600 ND ND ND 2.0 ND
14-18 y 2,800 1,800 50 800 ND ND ND 30 80 800 ND ND ND 3.0 ND
19-70 y 3,000 2,000 50 1,000 ND ND ND 35 100 1,000 ND ND ND 3.5 ND
> 70 y 3,000 2,000 50 1,000 ND ND ND 35 100 1,000 ND ND ND 3.5 ND
Pregnancy
≤ 18 y 2,800 1,800 50 800 ND ND ND 30 80 800 ND ND ND 3.0 ND
19-50 y 3,000 2,000 50 1,000 ND ND ND 35 100 1,000 ND ND ND 3.5 ND
Lactation
≤ 18 y 2,800 1,800 50 800 ND ND ND 30 80 800 ND ND ND 3.0 ND
19-50 y 3,000 2,000 50 1,000 ND ND ND 35 100 1,000 ND ND ND 3.5 ND
a UL = The maximum level of daily nutrient intake that is likely to pose no risk of adverse effects. Unless otherwise specified, the UL represents total intake from food, water, and supplements. Due to lack of suitable data, ULs could not be established for vitamin K, thiamin, riboflavin, vitamin B 12 , pantothenic acid, biotin, or carotenoids. In the absence of ULs, extra caution may be warranted in consuming levels above recommended intakes.b As preformed vitamin A only.c As ∂-tocopherol; applies to any form of supplemental ∂-tocopherol.d The ULs for vitamin E, niacin, and folate apply to synthetic forms obtained from supplements, fortified foods, or a combination of the two.e ß-Carotene supplements are advised only to serve as a provitamin A source for individuals at risk of vitamin A deficiency.f ND = Not determinable due to lack of data of adverse effects in this age group and concern with regard to lack of ability to handle excess amounts. Source of intake should be from food only to prevent high levels of intake.SOURCES : Dietary Reference Intakes for Calcium, Phosphorous, Magnesium, Vitamin D, and Fluoride (1997); Dietary Reference Intakes for Thiamin, Riboflavin, Niacin, Vitamin B 6 , Folate, Vitamin B 12 , Pantothenic Acid, Biotin, and Choline (1998); Dietary Reference Intakes for Vitamin C, Vitamin E, Selenium, and Carotenoids (2000); and Dietary Reference Intakes for Vitamin A, Vitamin K, Arsenic, Boron, Chromium, Copper, Iodine, Iron, Manganese, Molybdenum, Nickel, Silicon, Vanadium, and Zinc (2001). These reports may be accessed via www.nap.edu.Copyright 2001 by the National Academy of Sciences. All rights reserved. 2/15/01
Table 18. Dietary Reference Intakes (DRIs): Tolerable Upper Intake Levels (UL a ), Elements (130)Food and Nutrition Board, Institute of Medicine, National Academies
Life Stage Group Arsenic b Boron Calcium Chrom-ium Copper Fluoride Iodine Iron Magn-esium Manga-nese Molyb-denum Nickel Phos-phorus Selenium Silicon d Van-adium Zinc
--- (mg/d) (g/d) --- (µg/d) (mg/d) (µg/d) (mg/d) (mg/d) c (mg/d) (µg/d) (mg/d) (g/d) (µg/d) --- (mg/d) e (mg/d)
Infants
0-6 mo ND f ND ND ND ND 0.7 ND 40 ND ND ND ND ND 45 ND ND 4
7-12 mo ND ND ND ND ND 0.9 ND 40 ND ND ND ND ND 60 ND ND 5
Children
1-3 y ND 3 2.5 ND 1,000 1.3 200 40 65 2 300 0.2 3 90 ND ND 7
4-8 y ND 6 2.5 ND 3,000 2.2 300 40 110 3 600 0.3 3 150 ND ND 12
Males, Females
9-13 y ND 11 2.5 ND 5,000 10 600 40 350 6 1,100 0.6 4 280 ND ND 23
14-18 y ND 17 2.5 ND 8,000 10 900 45 350 9 1,700 1.0 4 400 ND ND 34
19-70 y ND 20 2.5 ND 10,000 10 1,100 45 350 11 2,000 1.0 4 400 ND 1.8 40
> 70 y ND 20 2.5 ND 10,000 10 1,100 45 350 11 2,000 1.0 3 400 ND 1.8 40
Pregnancy
≤ 18 y ND 17 2.5 ND 8,000 10 900 45 350 9 1,700 1.0 3.5 400 ND ND 34
19-50 y ND 20 2.5 ND 10,000 10 1,100 45 350 11 2,000 1.0 3.5 400 ND ND 40
Lactation
≤ 18 y ND 17 2.5 ND 8,000 10 900 45 350 9 1,700 1.0 4 400 ND ND 34
19-50 y ND 20 2.5 ND 10,000 10 1,100 45 350 11 2,000 1.0 4 400 ND ND 40
a UL = The maximum level of daily nutrient intake that is likely to pose no risk of adverse effects. Unless otherwise specified, the UL represents total intake from food, water, and supplements. Due to lack of suitable data, ULs could not be established for arsenic, chromium, and silicon. In the absence of ULs, extra caution may be warranted in consuming levels above recommended intakes.b Although the UL was not determined for arsenic, there is no justification for adding arsenic to food or supplements.c The ULs for magnesium represent intake from a pharmacological agent only and do not include intake from food and water.d Although silicon has not been shown to cause adverse effects in humans, there is no justification for adding silicon to supplements.e Although vanadium in food has not been shown to cause adverse effects in humans, there is no justification for adding vanadium to food and vanadium supplements should be used with caution. The UL is based on adverse effects in laboratory animals and this data could be used to set a UL for adults but not children and adolescents.f ND = Not determinable due to lack of data of adverse effects in this age group and concern with regard to lack of ability to handle excess amounts. Source of intake should be from food only to prevent high levels of intake.SOURCES : Dietary Reference Intakes for Calcium, Phosphorous, Magnesium, Vitamin D, and Fluoride (1997); Dietary Reference Intakes for Thiamin, Riboflavin, Niacin, Vitamin B 6 , Folate, Vitamin B 12 , Pantothenic Acid, Biotin, and Choline (1998); Dietary Reference Intakes for Vitamin C, Vitamin E, Selenium, and Carotenoids (2000); and Dietary Reference Intakes for Vitamin A, Vitamin K, Arsenic, Boron, Chromium, Copper, Iodine, Iron, Manganese, Molybdenum, Nickel, Silicon, Vanadium, and Zinc (2001). These reports may be accessed via www.nap.edu.Copyright 2001 by the National Academy of Sciences. All rights reserved. 2/15/01

9.7.2 Calcium Supplementation, Dietary Dairy Intake, and Weight Loss

Some studies in the past have suggested that calcium supplementation and/or supplementation of dairy products in the diet play a direct role in the prevention and treatment of obesity. However, not all data support this hypothesis, and several studies found that calcium or dairy consumption do not aid in weight loss (170), nor does calcium supplementation have an effect in preventing weight gain (171-173).

In a review evaluating 49 randomized clinical trials assessing the effect of dairy product or calcium supplement consumption, 41 studies showed no effect, two reported weight gain, one showed a lower rate of gain, and five showed it was effective as an aide in weight loss (170).

Major et al found that calcium plus vitamin D supplementation enhanced the beneficial effect of weight loss on the lipid profile; however, it had no effect on weight itself (174). There is also some evidence that high calcium or high dairy intakes during weight loss spare lean tissue loss to a greater extent than lower levels, although the evidence is not conclusive at this time.

 

9.7.3 Dietary Supplements and Weight Loss

Currently over half of the adult population uses dietary supplements (175). Most reported the motivation for using them was to increase over health even though less than a quarter of those supplements taken were recommended by a physician or healthcare provider (175). Retail sales of weight-loss supplements were estimated to be over $25.177 billion in 2008, including meal replacements (176).

Table 19.Common Ingredients in Weight Loss Dietary Supplements* (294)
Ingredient Purported Mechanism Research Findings Safety+
Bitter orange (synephrine) Increased energy expenditure and lipolysis; mild appetite suppressant Small clinical trials of poor methodological quality demonstrating possible effect on resting metabolic rate and energy expenditure, with inconclusive effects on weight loss Reported adverse effects include chest pain, anxiety, and increased blood pressure and heart rate
Caffeine (as added caffeine or from guarana, kola nut, yerba mate, or other herbs) Stimulation of central nervous system; increased thermogenesis and fat oxidation Short-term clinical trials of combination products showing possible modest effect on body weight or decreased weight gain over time Safety concerns not usually reported at doses less than 400 mg/day for adults, but there are significant safety concerns at higher doses.
Reported adverse effects include nervousness, jitteriness, vomiting, and tachycardia
Calcium Increased lipolysis and fat accumulation; decreased fat absorption Several large clinical trials have shown no effect on body weight, weight loss, or prevention of weight gain No safety concerns reported at recommended intakes, but constipation, kidney stones, and interference with zinc and iron absorption can occur at intakes above 2,000–2,500 mg for adults
Chitosan Binding of dietary fat in the digestive tract Small clinical trials, mostly of poor methodological quality, have shown minimal effect on body weight Reported adverse effects include flatulence, bloating, constipation, indigestion, nausea, and heartburn
Chromium Increased lean muscle mass; promotion of fat loss; reduced food intake, hunger levels, and fat cravings Several clinical trials of varying methodological quality have found minimal effect on body weight and body fat Reported adverse effects include headache, watery stools, constipation, weakness, vertigo, nausea, vomiting, and urticaria (hives) when taken above recommended intakes (25–45 mcg/day for adults)
Coleus forskohlii (forskolin) Enhanced lipolysis; reduced appetite A small number of clinical trials show no effect on body weight Unknown
Conjugated linoleic acid Promotion of apoptosis in adipose tissue Several clinical trials have shown minimal effect on body weight and body fat Reported adverse effects include abdominal discomfort and pain, constipation, diarrhea, loose stools, dyspepsia, and possible adverse effects on blood lipid profiles
Ephedra (ma huang, ephedrine) Stimulation central nervous system; increased thermogenesis; reduced appetite Several short-term clinical trials of good methodological quality, many of ephedra combined with caffeine, have found modest effect on short-term weight loss Banned as a dietary supplement ingredientReported adverse effects include anxiety, mood changes, nausea, vomiting, hypertension, palpitation, stroke, seizures, heart attack, and death
Garcinia cambogia (hydroxycitric acid) Inhibited lipogenesis; suppressed food intake Several short-term clinical trials of varying methodological quality have found little to no effect on body weight Reported adverse effects include headache, nausea, upper respiratory tract symptoms, and gastrointestinal symptoms
Glucomannan Increased feelings of satiety and fullness; prolonged gastric emptying time Several clinical trials of varying methodological quality, mostly focused on effects on lipid and blood glucose levels, have found little to no effect on body weight Tablet forms may cause esophageal obstructions. Other reported adverse effects include loose stools, flatulence, diarrhea, constipation, and abdominal discomfort
Green coffee bean extract (Coffea aribica, Coffea canephora, Coffea robusta) Inhibited fat accumulation; modulated glucose metabolism Few clinical trials of poor methodological quality have suggested possible modest effect on body weight Reported adverse effects include headache and urinary tract infections
Green tea (Camellia sinensis) and green tea extract Increased energy expenditure and fat oxidation; reduced lipogenesis and fat absorption Several clinical trials of good methodological quality on green tea catechins with and without caffeine have shown possible modest effect on body weight Reported adverse effects for green tea extract include constipation, abdominal discomfort, nausea, increased blood pressure, liver damage
Guar gum Bulking agent in gut; delayed gastric emptying; increased feelings of satiety Several clinical trials of good methodological quality have found no effect on body weight Reported adverse effects include abdominal pain, flatulence, diarrhea, nausea, and cramps
Hoodia (Hoodia gordonii) Suppressed appetite; reduced food intake Very little published research in humans, but results from one study suggest no effect on energy intake or body weight Concern for increased heart rate and blood pressure. Other reported adverse effects include headache, dizziness, nausea, and vomiting
Pyruvate Increased lipolysis and energy expenditure Few clinical trials of weak methodological quality suggest possible minimal effect on body weight and body fat Reported adverse effects include diarrhea, gas, bloating, and possible decreases in high-density lipoprotein levels
Raspberry ketone Altered lipid metabolism Studied only in combination with other ingredients. Unable to draw conclusions. Unknown
White kidney bean (Phaseolus vulgaris) Acts as a “starch blocker, interfering with breakdown and absorption of carbohydrates Few clinical trials, all of poor methodological quality, suggest
possible modest effect on body fat, but no effect on body weight
Reported adverse effects include headache, soft stools, flatulence, and constipation
Yohimbe (Pausinystalia yohimbe, yohimbine) Hyperadrenergic effects Very little research has been done on yohimbe for weight-loss, with insufficient evidence to draw firm conclusions. Significant safety concerns reported, with adverse effects including headache, anxiety, agitation, hypertension, and tachycardia
*Table adapted from The ODS Fact Sheet on Dietary Supplements for Weight Loss (294)
+Listed in order of severity, with the most severe reported side effects listed last.

In 2001, MetaboLife® 356, an Ephedra-containing combination supplement, was the top selling dietary supplement. It
reached $70 million in sales, but it was also responsible for 64 percent of all herb-related adverse events reported to the U.S. Poison Control Center during that same year (175;177). Ephedra, or Ma Huang, is the common name for the herb that was used in many of these weight loss supplements. It is an herb used in traditional Chinese Medicine (TCM). Its use in weight reduction though, is not a common practice in TCM. Americans used this supplement as a weight loss aid from the mid 1990’s, up until 2004, when it was banned by the FDA (178;179).

The NIH sponsored a thorough systematic review of the safety and efficacy of Ephedra through the Agency of Healthcare Research and Quality’s (AHRQ) Evidence Based Practice Center at the University of Southern California, which conducted the study. It concluded that the use of Ephedra, with or without caffeine, correlated with a small but nonetheless statistically significant increase in weight loss over six months, (almost equal to 0.9 kilograms per month more than with the placebo). The weight lost by those taking Ephedra in combination with caffeine exceeded weight lost by prescription medications in two head-to-head randomized, double-blinded clinical trials (178). There were no studies that measured the long-term effects (more than 6 months) of Ephedra use, and the problem was that the supplement was not safe. Adverse effects of the supplement in the AHRQ study included two to three times more nausea, vomiting, psychiatric symptoms such as anxiety and change in mood, autonomic hyperactivity, and palpitations when compared with placebo. Serious adverse events (SAE’s) were defined as specified by FDA criteria. SAEs were reported to the FDA, and adverse event reports from a manufacturer of Ephedra-containing dietary supplements were also evaluated in the RAND/Southern California systemic review. These reports raised concern about the safety of dietary supplements containing Ephedra due to the number of deaths, myocardial infarctions, cerebrovascular accidents, seizures, and serious psychiatric illnesses in young adults, data was sufficient to warrant concern (178).

The FDA concluded in 2004 that Ephedra-containing products were not to be recommended for weight loss. There was unreasonable risk for illness and injury when taking such dietary supplements. Thus, the sale of dietary supplements containing Ephedra has been prohibited in the United States since April 2004 (178). Ephedra like supplements such as Citrus aurantium (Bitter orange) may also pose risk.

The latest information on dietary supplements and weight loss can be found at www.ods.nih.gov.

 

9.8 Fiber

The next section outlines dietary fiber needs during weight reduction.

9.8.1 Fiber Needs in Reducing Diets

Dietary fiber is chemically similar to carbohydrate in most of its forms but it is virtually non-caloric because the human body lacks the enzymes to break the fiber’s glycosidic bonds. Some short-term experimental and several cross-sectional studies suggest that an increased dietary fiber intake reduces weight gain. In contrast, fiber is not effective as a weight loss aid (180). Fiber should be included in reducing diets at levels of about 25 to 38 grams per day to facilitate laxation. Both soluble and insoluble dietary fiber may also modify hunger and help to sustain satiety, but again experimental evidence is not conclusive (10;181;182). Inclusion of five or more servings of fruits and vegetables daily, with plenty of whole grain breads and cereals helps to meet both soluble and insoluble fiber goals on reducing diets. On a VLCD, it is also important to include at least some fiber. As dietary fiber intakes increase, water requirements also increase, so intakes of fluid should also be substantial. Adequate fiber and water are essential for maintaining a soft stool and normal laxation. Ample fiber intakes are associated with reduced risk of several chronic diseases (183).

9.9 Energy-Dense Beverages, Alcohol, and Energy-Free Artificially and Naturally Sweetened Beverages

Currently, it is estimated that the mean intake of ”added” sugars in the American diet is about 15.8% of total energy, and that the largest source of these added sugars is from calorically sweetened beverages such as soft drinks, fruit aides, and other sweetened beverages, accounting for 47% of total added sugars in the diet (184;185) . The term “energy-dense beverage” encompasses a wide variety of beverages, including sugar sweetened soda, fruit drinks, juice, lemonade, sweetened iced tea, milk and soy beverages, and alcohol. Alcohol is especially calorie dense (7 Cal/gm, vs 4 for sugars), and it also may bypass satiety mechanisms and lead to a lack of ability to control eating. Curiously, in recent years it has been neglected in spite of the fact that many adults (and unfortunately some younger people) drink alcohol on a regular basis.

9.9.1 Energy-Dense Beverages

When compared to water or energy-free beverages, consumption of energy-containing beverages tends to increase total energy intake from meals (184). Although the evidence is not highly conclusive, it is argued that: 1) consumption of an isocaloric beverage compared to consumption of solid food prior to a meal increases food intake, 2) solid foods enhance satiety hormones more than energy-dense beverages, and 3) energy-dense beverages are often comprised largely of refined carbohydrates, which stimulate fewer satiety signals than unprocessed carbohydrates, fat or protein (184). More research is needed on this issue to validate each of these assumptions. It should be noted that alcohol is also energy dense and bypasses satiety mechanisms. This is discussed below further in section 9.9.2 Alcohol.

The Beverage Guidance Panel recommends that the average person limit daily consumption of caloric, sweetened beverages without nutritional benefits (soft drinks, fruit drinks, fruit cocktails, fruit aids, and sweetened teas and coffees) to eight ounces per day. Caloric, nutrient-dense beverage consumption (milk, soy and 100% fruit and vegetable juices) should be kept to a minimum (186). In the United States, a regular, 12-ounce can of calorically sweetened soda provides approximately 150 calories, typically in the form of high-fructose corn syrup. These calories, if not balanced with exercise or a caloric reduction in other areas of one’s diet, could gradually lead weight gain over time (187).

While there may be health benefits to consuming energy and nutrient dense beverages such as milk and 100% fruit juice, the additional energy provided by these energy-dense beverages must be offset by an increase in energy expenditure or a decrease in other areas of energy consumption in order for weight loss or weight maintenance goals to be achieved. Any energy-dense beverages included in the diet should also be nutrient dense, such as various forms of milks (e.g. diary, soy) and 100% fruit juices (which should be limited to about one serving per day). Most other caloric beverages, such as sodas, fruit drinks, sweetened coffees and teas provide calories with little or no vitamins, minerals, or other nutrients. For this reason, energy-dense beverages are generally not recommended for patients attempting to lose weight.

 

9.9.2 Alcohol

Alcohol (ethanol) contains approximately seven calories per gram, providing more energy per unit of weight than either carbohydrate or protein (each providing about four calories per gram), but less than fat (about 9 calories per gram). Alcoholic beverages are a source of non-nutritive energy, or “empty calories”. If protein, carbohydrate and/or fat are consumed at the same time as alcohol is ingested, their oxidation will be suppressed (most notably fat oxidation), since alcohol is preferentially oxidized, and the other macronutrients balance through the sparing effect of alcohol on fat oxidation. This may lead to increased fat storage.

In addition to alcohol’s influence on macronutrient metabolism, chronic extremely excess intake of alcohol also interferes with the absorption and utilization of several vitamins and minerals. Alcohol in excess also impairs nutrient absorption by damaging the stomach and intestinal lining, disabling the transport of some nutrients into the blood. Chronic overconsumption of alcohol can also lead to fatty liver, dyslipidemia, and further weight gain, and should be discouraged.

Another important consideration concerning alcohol’s influence on energy balance is its effects on energy intake. Alcohol is positioned at the bottom of the hierarchy of satiating efficiency of metabolic fuels consumed by humans (10). Generally, satiety provided by fuels is ranked from lowest to highest: alcohol, fat, carbohydrate (depending on type), and protein (188). Alcohol energy is additive to the diet, producing no compensation in energy intake under most ad-libitum situations, and in fact, some research suggests that alcohol may stimulate appetite (189;190). For these reasons, alcohol consumption is usually contraindicated on weight-loss diets.

 

9.9.3 Low and No Calorie Sweeteners

These ingredients are variously called low and no calorie sweeteners, non-nutritive sweeteners, sugar substitutes, reduced calorie sweeteners, or artificial sweeteners). They are added to foods and beverages to provide sweetness without adding significant amounts of sugar and calories to the product. They are also used as flavorings to mask the bitter taste of drugs http://www.caloriecontrol.org/sweeteners-and-lite/sugar-substitutes.

Low calorie sweeteners are appealing because they satisfy our innate preference for sweetness without the associated calories consuming sugar would have. They do not contribute to dental caries, may help to make low calorie diets more palatable, and assist compliance. These low calorie sweeteners are consumed by approximately one fifth of U.S. adults (191). In human studies, data have been conflicting on whether they are associated with weight loss (192). However, the draft conclusions of the 2015 Dietary Guidelines Scientific Advisory Committee concluded that there was moderate and generally consistent evidence from randomized clinical trials conducted in adults and children supporting the contention that replacement of sugar-containing sweeteners with low calorie sweeteners reduces calorie intake, body weight and adiposity. However evidence was judged only limited from long-term observational studies in children and adults that there was an association between low calorie sweeteners and body weight compared to sugar containing sweeteners. The Committee further concluded in its draft statement that there was only limited long-term observational study evidence in adults that there was an association between low calorie sweeteners and risk of type 2 diabetes.

The FDA has approved seven non-nutritive sweeteners for use in foods and beverages in the United States to date. Most are regulated as food additives, including acesulfame-potassium (Acesufame-K. Ace-K, Sunett, Sweet One), aspartame (Equal, NutraSweet), saccharin (Necta Sweet, Sugar Twin, Sweet ‘N Low), sucralose (Splenda), and two products that are used largely as ingredients rather than as table top sweeteners, neotame, and most recently, advantame. Cylamate is not sold in the United States but is sold in many other countries as a tabletop sweetener. The FDA granted stevia leaf, often called “sweet leaf”, a plant-derived sweetener, (PureVia, Sun Crystals, and Truvia Generally Recognized As Safe (GRAS) status. It is derived from stevia leaves by steeping them in water and purifying to extract to obtain only high purity rebaudioside A. The ingredient is sold under the trade name Rebiana and is the major source of sweetness in the Truvia sweetener brand. , Also GRAS is the product Fruit-Sweetness™ is anon-caloric fruit concentrate sweetener derived from the monk fruit, a traditional fruit originating in Southeast Asia (193). The sweeteners have a variety of different chemical structures, as shown in Table 20, and summarized elsewhere (http://www.caloriecontrol.org/sweeteners-and-lite/sugar-substitutes).They also vary widely in their sweetness and in their most common uses. The most commonly consumed source of these products is as a sweetener to beverages either provided by sachets of the different products, that are added to the drink or as an ingredient in a variety of “diet’ soft drinks and colas. The sweeteners used in foods and beverages are listed on the ingredient list for those who prefer a specific ingredient.

There is little evidence that low calorie sweeteners in and of themselves can cause patients to lose weight; their use must always be coupled with a hypocaloric diet. The low calorie sweeteners may make reducing diets more palatable and encourage compliance, but this remains to be demonstrated conclusively (194-195). During weight maintenance, water and other non-caloric beverages or, low calorie sweetened beverages are a preferable alternative to high calorie beverages, such as regular soft drinks, sweet teas, other sugary drinks and alcohol. Some recommend that adults consume no more than 32 ounces of low calorie sweetened beverages per day, but there is little evidence supporting such recommendations (186). If an individual wishes to reduce his/her exposure to a particular low calorie sweetener, both consuming less or choosing a variety of low calorie sweeteners are useful strategies to consider

9.9.3.1 SAFETY OF LOW CALORIE SWEETENERS

The safety of low-calorie sweeteners has been evaluated by several bodies, including the US Food and Drug Administration (FDA), the Joint Expert Committee of Food Additions (JECFA) of the United Nations Food and Agricultural Organization (FAO), the World Health Organization (WHO), the Scientific Committee on Food (SCF) of the European Commission, and the European Food Safety Authority (EFSA) (242, 254). In the United States, the use of sweeteners is regulated by FDA under the 1958 Food Additives Amendment of the Food, Drug and Cosmetic Act of 1938 (249). Currently, two low calorie nutritive sweeteners sugar alcohols (mannitol and xylitol) and six non-nutritive sweeteners (aspartame, acesulfame-K, saccharin, sucralose, neotame, advantame) are approved as food additives by the FDA (242), and two additional “natural” sweeteners, extracts of stevia and monk fruit, are approved as GRAS.

An Acceptable Daily Intake (ADI) is established for all FDA-approved low-calorie sweeteners. It represents the amount of each sweetener that can be safely ingested daily by a human over a lifetime without risk based on animal toxicity studies. The ADI is expressed as milligrams per kilogram of body weight. The ADI is not a maximum intake level. It is a conservative estimate of the level-- a hundredth of the maximum level-- at which no observed adverse effect is seen in feeding studies of toxicity in experimental animals (243). Consumption of these low-calorie sweeteners at levels below the ADI is considered by the FDA to be safe for the entire population, including children and pregnant women (246). Exceptions for particularly vulnerable populations, such as individuals who suffer from phenylketonuria (a rare metallic disorder), who should not consume aspartame-sweetened products, are listed in the table, and on the product label.

Low-calorie sweeteners such as stevia and monk fruit extracts that are GRAS are also are acceptable only for specified uses in specified amounts that are in line with traditional uses of these ingredients in human diets over the course of history. Generally Recognized as Safe (GRAS); substances are those that scientific experts have agreed are safe for use in appropriate amounts in foods and beverages, based largely on traditional use. They do not require the extensive testing required by FDA for food additives, but are permitted to be used only at specified levels (244). Also, some other compounds with sweet tastes, including thaumatin, neohesperidine, and glycyrrhizin are GRAS when used in small amounts for an intended use as flavor enhancers, but not as sweeteners.

9.9.3.2. Low calorie sweeteners that are currently approved by FDA:

Saccharin—Sold under the brand names Sweet ‘N Low®, Sugar Twin®, and Sweet Twin®, saccharin is a non-nutritive sweetener that is not metabolized by the body and provides no calories. It has been on the market longer than any other low calorie sweetener. It is 300-500 times sweeter than sugar and is heat stable (246), but leaves a bitter metallic aftertaste in the mouth in individuals who have a certain dominant genetic trait. They can be identified by the ability to taste the chemicals ptc (phenylthiocarbamide) and PROP (6 propyl 2 thiouracil) as bitter or metallic. Those compounds do not appear in food but related compounds such as saccharine and possibly some of the Brassica vegetables have related compounds that are also tasted as bitter. About 70 % of Americans taste ptc related compounds in food as bitter, especially if they are not habituated to coffee or tea or non-smokers, and therefore in the USA saccharine is usually blended with other sweeteners in food and beverage products (252). Many Asians are very sensitive to a bitter taste, and formulations of low calorie soft drinks for them may use other sweeteners (247). The ADI for saccharin is 15 mg/kg/day. Studies in the 1970s in rats linked a lifetime exposure to high doses of saccharin to development of bladder cancer. This raised concerns about the safety of saccharin and led to a ban of saccharin in 1977 by the FDA, and great furor since at the time there were few other non-nutritive sweeteners on the market. Later, after the ban was lifted due to Congressional action, FDA required that products containing saccharin must carry a warning label (244). Subsequent human case- control and other studies showed no association between saccharin and bladder cancer development (254). In addition, it was later shown that physiological differences in bladder and urine chemistry between rats and humans were such that the bladder cancer-causing effect was specific only to rats, and that the toxic effect of tumors in rats was evident only with sodium saccharin but not with other forms of saccharin, and only with lifetime exposures to very high dosages of saccharin, equivalent to hundreds of servings per day in humans (247). After these studies were reviewed, in 2000 saccharin was delisted as a potential human carcinogen. However, products containing saccharin must still label saccharin in declarations on their ingredient list. Today, several professional societies, including the American Dietetic Association, American Medical Association, and American Cancer Society state that saccharin is safe and acceptable for use in all populations (242).

Acesulfame K (Ace-K)—Under the brand names of Sunett® and SweetOne®, Ace-K is a combination of an organic acid and potassium. It is excreted from the body unchanged, and therefore does not yield either a net increase in calories or of potassium in humans. It is 200 times sweeter than sugar. It is heat stable, and thus is suitable for cooking and baking (242). Ace-K also leaves a metallic aftertaste in some people’s mouths when it is used by itself, and so it is usually combined with other sweeteners in sweetener blends, especially in carbonated beverages. The ADI for Ace-K is 15 mg/kg/day; for a person of 70 kg that would be about 1050 mg. The typical amount of Ace-K in a 12-oz beverage is 40 mg and in a packet of tabletop sweetener it is 50 mg (257). Long-term human studies found no effects on cancer even at very high consumption levels of Ace-K. There are also no case reports documenting adverse health effects associated with Ace-K. Therefore, FDA permits Ace-K use in all population segments. An Ace-K metabolite, acetoacetamide, is toxic when consumed at very high doses, but the amount of acetoacetamide found in beverages sweetened with Ace-K is negligible. And therefore consumption of beverages sweetened with Ace-K is deemed to be safe (247).

Sucralose—Sucralose is sold under the trade name of Splenda®. It is a sugar derivative, which replaces 3 hydrogen-oxygen groups on the sugar with 3 chlorine atoms. Although sucralose is derived from sugar, it is not absorbed nor do gut enzymes in humans metabolize sucralose as a carbohydrate. It is non-caloric (251). It is 600 times sweeter than sugar and is highly heat stable. Sucralose is used extensively in foods and beverages because it retains sweetness over a wide range of temperatures and storage conditions (242). The ADI for sucralose is 5 mg/kg/day. Sucralose had no significant effect on blood glucose control in individuals with type 2 diabetes mellitus even at levels 3 times this amount over three months. Extensive research in humans and experimental animals has found no association between sucralose consumption and carcinogenicity or other health concerns (244). Therefore, sucralose appears to be safe and acceptable for human consumption.

Aspartame—It is sold under the trade names NutraSweet®, Canderel®, Sanecta®, TriSweet®, E951 (for food ingredients), and Equal® (for the tabletop sweetener). Aspartame is a synthetic sweetener composed of aspartic acid and phenylalanine (256). It is used both as an ingredient and sold to consumers as a tabletop sweetener. Since it is metabolized to amino acids, aspartame provides 4 calories per gram. It is 160-220 times sweeter than sugar. Due to its intense sweetness, only a tiny amount (e.g. a few mg) is needed to sweeten a food and thus, the energy provided from the compounds is negligible. Aspartame decomposes and loses its sweetness with heat and so it is not suitable for baking and cooking. However, it is used extensively in soft drinks, which account for more than 70% of aspartame consumption in the US (242). An 8-oz diet Coca Cola contains approximately 125 mg of aspartame and 60 mg of phenylalanine (257). The ADI of aspartame is 50 mg/kg/day (or about 3500 mg for a 70 kg individual). Due to the presence of phenylalanine in aspartame, individuals with phenylketonuria (PKU), a recessive inborn error of metabolism should be cautioned about aspartame consumption since they cannot metabolize phenylalanine effectively and the amino acid can accumulate in the blood, causing toxic metabolites and potentially neurological damage (242). Although this adverse effect is unlikely because the amounts of aspartame that are used in foods and as a sweetener is quite small, diet therapy for PKU includes limiting dietary phenylalanine, including aspartame. FDA requires products containing aspartame to carry the warning label “PHENYLKETOURICS: CONTAINS PHENYLALANINE.” The plasma response of phenylalanine to ingestion of aspartame varies in people with PKU, but most seem to tolerate the amount of phenylalanine in a single diet soda sweetened with aspartame (about 104 mg phenylalanine/12-oz can), so an immediate health crisis is unlikely should a person mistakenly drink a diet soda (244). However, caution is still indicated for PKU patients. In contrast, healthy adults show no significant change in plasma level of aspartic acid even with doses of aspartame about 4 times the ADI (4 times ADI is equal to 200 mg/kg/day) (256). In normal humans, plasma phenylalanine increases in a dose-response manner to aspartame dosage in the range of 2-100 mg/kg/day without any observed effects on cognitive function, but individuals with phenylketonuria are different and may be particularly sensitive (258).

Aspartame is perhaps the most controversial sweetener of them all due to early claims in the 1970S of neurotoxicity in primates, which were later not confirmed, although the unproven claims continue to circulate on the Internet. One comprehensive review study on the safety of low-calorie sweeteners, including aspartame, stated that “much of potential misinformation about aspartame and health seems to be based on misunderstandings or partial scientific truths” (247) some consumers report adverse reactions to aspartame, including headache, facial edema, skin reactions, respiratory problems, seizures, and behavioral and cognitive changes, numerous controlled studies have failed to reproduce these adverse reactions reported. Adding to the controversy, aspartame was claimed by one Italian laboratory to be carcinogenic, but no associations between aspartame consumption and cancer development were found in a review of their data by a panel of experts in the experimental studies in rodents upon which the claim was based, and the studies themselves were criticized as being inadequate (256) Other data reviewed also were negative for adverse effects. The National Cancer Institute at NIH concluded that there was a lack of association between aspartame consumption and increased cancer risk, even at high intake levels after renewing all of the data (259). The European Food Safety Authority (EFSA) panel in 2009 again reviewed all available scientific evidence on the safety of aspartame and concluded that there was no carcinogenic effect nor association with neurobehavioral disorders or other effects from it and that no further revision of the ADI was needed. The most recent statement of EFSA released in 2013 has also confirmed the safety of aspartame (260). Periodic updates will be made available. Current evidence suggests that aspartame is safe at levels below ADI other than for individuals with PKU, and there is no credible evidence showing carcinogenicity, neurotoxicity, and other adverse health outcomes even at levels above ADI.

Neotame—Neotame is a relatively new non-nutritive low calorie sweetener, approved by FDA in 2002. It is used as an ingredient and also as a flavor enhancer that can modify other flavors in foods and beverages (246). Neotame is chemically related to aspartame, being composed of aspartic acid, methanol, and phenylalanine. Although they are similar in chemical structure, neotame and aspartame are completely different compounds with different physical and biological properties (247). Neotame is 7000-13000 times sweeter than sugar, thus only an infinitesimal amount is required for sweetening. The ADI for neotame is 2 mg/kg/day in other countries and 18 mg/day in the United States. Although neotame is metabolized into phenylalanine, a PKU warning label is not required for products containing neotame because the amount of neotame to sweeten a food is so tiny, owing to its intense sweetness. Thus, exposure to phenylalanine from neotame is negligible and clinically insignificant (244). In addition, neotame is not directly metabolized to phenylalanine (252). The blockage of peptidases that break the peptide bond between aspartic acid and phenylalanine decreases the availability of phenylalanine in the bloodstream after neotame ingestion. Some concerns have been raised regarding neotame being neurotoxic due to the structural similarity to aspartame. However, no adverse effects of this sort or of other concerns have been found (258). Therefore, neotame is considered safe and acceptable for use in all populations.

Advantame This ingredient is an intense low calorie non-nutritive sweetener approved in 2014. It is used chiefly as a food ingredient, and is not sold over the counter. It was approved in 2014 for use in foods and beverages; USDA must approve uses in meat and poultry as well. It is structurally somewhat similar to aspartame (295).

Stevia (Stevioside, Steviol glycosides, Rebaudioside A)—These are sold under the trade names Truvia®, Sun Crystals®, PureVia®, Sweetleaf Sweetener®, Stevia in the Raw®, and Enliten®. Stevia leaves, often called “sweet leaf”, have been used for centuries in Asian and South American countries as non-caloric sweetener and traditional medicine (261). Steviol glycoside is the sweet component derived form leaves of Stevia rebaudiana. It is 250-300 times sweeter than sugar. Stevioside extracts have been used for sweetening in pickled vegetables, seafoods, soy sauce, soft drinks, and confectionary (262). Stevia leaves have also been used medicinally in Asia and South Africa for hypertension, hyperglycemia, obesity, and skin disorders, although the evidence for beneficial effects is scanty and none of these therapeutic uses are approved in the US. Stevioside is not metabolized nor absorbed by the digestive tract. The use of steviosides as a food ingredient was not initially approved in the United States although it was in Europe. Rather, it was sold in the US initially as a dietary supplement and therefore it could not be marketed as a sweetener since this was a different intended use. In 2008, FDA granted stevia GRAS status for use as a general-purpose sweetener in addition to use as a dietary supplement (263). Stevioside can now be found as an ingredient in beverages and as tabletop sweeteners. The Joint Expert Committee on Food Additives (JECFA) conducted a thorough scientific review on stevioside and concluded that it was safe for use in food and beverages with no major toxicity risk. The ADI of 0-4 mg/kg/day has recently been established by JECFA (296).

9.9.3.2 Nutritive low calorie sweeteners

Polyols (Sugar alcohols)—Sugar alcohols (polyols) are considered nutritive sweeteners because they provide some calories in the diet. However, the amount of calories provided per gram is fewer than table sugar (about 2 vs 4 calories/gram) due to the alcohol’s incomplete digestion and absorption. The unabsorbed polyols reach the colon and cause subsequent fermentative degradation by intestinal bacteria (297, 248). Most polyols are about half as sweet as sugar. They replace sugar in the products for sweetness and volume, and therefore, products containing them can be labeled as “sugar-free”. Due to the osmotic effect of unabsorbed polyols in the large intestine, products containing polyols may cause bloating, gas, discomfort, and diarrhea when consumed in excess (e.g. > 50 g/day sorbitol and >20 g/day mannitol). Therefore, these products are required to carry the statement “Excess consumption may have a laxative effect” on the label. Sensitivities to this laxative effect differ among individuals. Children may be particularly sensitive to the laxative effects of polyols due to their smaller body size (244). Tolerance to polyols can be increased somewhat with a gradual increase of intake that allows for adaptation to their laxative effects (264). Polyols may also confer potential health benefits, such as reduction in dental caries, a lower glycemic response when they are used in place of sugar, provision of fewer calories than sugar, and possibly a prebiotic effect (265). The prebiotic effects are presently poorly documented.

There are three subtypes or categories of Polyols

Polyolnosaccharides:

1) Erythritol—Marketed under the brand name Zerose, erythritol is found naturally in pears, melons, grapes, and mushrooms. Due to its almost complete absorption and unchanged excretion in urine within 24 hours, erythritol may not have as potent a laxative effect as some of the other polyols. In addition, erythritol provides fewer calories per gram (0.2 calories per gram) than the other polyols (253, 264).

Sorbitol—Usually found in sugar-free candies, chewing gums, baked goods, frozen desserts, and toothpaste, sorbitol is non-cariogenic. It is also slowly absorbed in the gut and metabolized independently of insulin (248, 253). Thus, it may be beneficial for individuals with diabetes.

Mannitol—Extracted from seaweed, mannitol is poorly absorbed and may cause a stronger laxative effect at a relatively lower dose (10-20 gram) than other polyols (252, 253). It is used as a dusting powder for chewing gum, an ingredient in chocolate-flavored coating agents for ice cream and candies, and in pharmaceuticals. It also cools the mouth and masks bitter tastes.

Xylitol—Xylitol is extracted from birch, raspberry, plum, and corn. It is often found in sugar-free gums, cough drops, mints, and oral health products. Several clinical trials have shown that xylitol is more effective in reducing dental caries than any of the other polyols. In addition, xylitol may even have a caries-preventive and anti-cariogenic effect (4). Due to this, FDA authorizes the health claim that “xylitol does not promote tooth decay” on product labels (256).

2) Polyol Disaccharides:
Maltitol-- Derived from maltose, maltitol is a food ingredient that is metabolized into glucose and sorbitol by the gut flora. It is used in chocolate candies, jams, baked goods, and ice cream to give texture to products, and sometimes as a fat replacer (http://www.caloriecontrol.org/sweeteners-and-lite/polyols).

Lactitol –Although it is derived from lactose, lactitol is not hydrolyzed by lactase in small intestine. Instead, it is fermented by the microflora in the large intestine and converted into biomass and short-chain fatty acids. Also, it acts as a prebiotic, stimulating growth of the gut bacteria. The health effects of this are uncertain, but because lactitol is fermented in the colon, it may act as prebiotic and may produce more intestinal discomfort than some of the other products (249, 253). Lactitol is used as an ingredient in bakery products, hard and soft candies, frozen dairy desserts, and chocolate (248)

Isomalt—Under the trade name Palatinit, isomalt is incompletely metabolized into mannitol, sorbitol, and glucose in small intestine. It is noncariogenic and triggers only a low glycemic response (242). It is approved in most European countries and is added to candies, toffee, fudge, wafers, and cough drops. 90% of the isomalt is fermented further in colon, making it also a potential prebiotic (265). Intakes of 30 grams of isomalt might increase numbers of bifidobacteria in the colon, but evidence is still uncertain on this, and even if it did the effects of increased bifidobacteria on health are not yet well documented.

3) Polyol Polysaccharides:

Hydrogenated starch hydrolysate (HSH)—HSH is produced by the hydrolysis of corn, wheat, or potato starch. Lycasin®, Hystar®, Stabilite®, and Roquette's 75/400 are the HSH brands currently available on the market. They are used mostly as bulking agents and food ingredients to provide sweetness, volume, and texture in commercially produced “sugarless” food products. HSH is metabolized into maltitol, sorbitol, and glucose to provide 3 calories per gram (253). The glycemic index of HSH is similar to that of maltitol, and about 40% of it is digested in the intestine (264). Therefore, it may be suitable for individuals with diabetes but this claim needs more evidence to support it.

9.9.3.4 Other sweeteners pending approval:

These sweeteners are not yet fully approved by FDA for use in food:

Cyclamate—Sold under the trade names SugarTwin® and Sucaryl®, cyclamate is 30 times sweeter than sugar, provides no calories, and blends well with other sweeteners. Most individuals do not metabolize cyclamate (252). It is approved for use as a tabletop sweetener in more than 50 countries, but re-approval is currently held abeyance and pending in the United States. Cyclamate has been banned in the US since 1969 based on a study suggesting the association between a saccharin/cyclamate blend sweetener and bladder cancer in rats (266). Cyclamate is converted to its metabolite, cyclohexylamine, which is relatively toxic and may cause bladder cancer in rats. High doses of cyclohexylamine cause male infertility in rats, although this effect is not observed in humans (247). Studies since 1969 have found no relationship between cyclamate and bladder cancer in humans, and this is the evidence that is currently being considered by FDA pending re-approval. FDA has stated that animal studies on mice and rats do not implicate cyclamate as a carcinogen.

Alitame – Under brand name Aclame®, alitame is derived from amino acids alanine and aspartic acid. Only the aspartic acid component is metabolized, to yield 1.4 calories per gram. Since only a minute amount of the sweetener is used as a food ingredient, the energy provided is negligible. It is 2000 times sweeter than sugar and heat stable. Common applications of alitame are commercially produced baked goods, candies, frozen desserts, beverages, and pasteurized foods. Alitame has been approved by several countries and is pending approval in the USA (242, 253).

Thaumatin—Sold under trade name Talin®, thaumatin is a protein sweetener that occurs naturally in a West African fruit Thaumatococcus danielli. It is 1600 times sweeter than sugar. However, it leaves a licorice-like aftertaste. Its application in food products is very limited due to the delayed appearance and extinction time of the sweet sensation and this licorice aftertaste (249). In the United States, thaumatin is recognized as GRAS as a flavor enhancer (especially in chewing gums), but not as a sweetener. The Joint Expert Committee on Food Additives (JECFA) reviewed the safety of thaumatin and found no toxicity. The ADI for thaumatin is not yet specified (244).

Neohesperidine dihydrochalcone – A derivative of bioflavonoids in citrus fruits, neohesperidin dihydrochalcone is 1500 times sweeter than sugar but has different flavor profile. It also leaves a licorice aftertaste. However, it intensifies the mouth feel of juices, and thus has the potential to be used in fruit juice, chewing gums, and mouthwash (242). It is metabolized by the gut flora. Although it is approved in the European countries, it is recognized as GRAS only in amounts for use as a flavor enhancer in the United States, but not for the larger amounts that would be needed for use as a sweetener

Glycyrrhizin—An extract from licorice root, glycyrrhizin is 30 times sweeter than sugar (247). It has limited use as a flavoring agent in some candies and tobacco due to its strong licorice flavor. It is also GRAS in the United States as a flavoring agent, flavor enhancer, and surfactant, but not as a sweetener (249).

9.9.3.5 POSSIBLE HEALTH BENEFITS OF LOW CALORIE SWEETENERS:

Weight loss and maintenance

At different times and places overindulgence of various foods has occurred. In the US today many people of all ages indulge in sugary sweet foods and beverages high in calories, and adults also in alcohol. Reduction of food energy from any source, including these sources, will result in weight loss over the long term if other foods are not substituted for it. In 2014 the Obesity Society issued a statement that reduced consumption of sugar-sweetened beverages can reduce total caloric intake and that individuals and especially children and those with weight problems reduce their consumption of sugar sweetened beverages (298). Low calorie sweeteners offer an option other than water for alternative beverages, but the reality remains that in order to lose weight, the dieter must achieve and maintain a negative energy balance (e.g. Energy intake < energy expenditure) without compensating for what is e decreased by increasing intakes of other foods or beverages. . Some randomized-controlled studies have shown beneficial effects of low calorie sweeteners in weight management; with a small short-term weight loss (of about 0.2 kg/week) over placebo with the low calorie sweeteners plus a hypocaloric diet, and improved weight maintenance after the weight loss diet is discontinued while others do not (242, 267, 268, 269, 270). Another study found that when the artificial sweetener group (particularly aspartame) was compared to the control it showed a difference of about 5 kg (maintained a weight loss of 5.1 kg longer), when artificial sweeteners were substituted for caloric sweeteners in beverages and with a hypocaloric diet (267). Other studies were not as positive. A review of aspartame’s role on weight control in 2006 showed a weight loss of 0.2 kg/week when products sweetened with sucrose was substituted with aspartame (269) in a hypocaloric diet. Another study showed more successful maintenance of weight loss after three years in women who were encouraged to consume aspartame-sweetened products (247).

In the most definitive study to date, of 303 participants who were overweight, during a 12 week weight loss program those who used low calorie sweetened beverages instead of water alone lost more weight than those in the control group (5.95kg vs 4.09 kg) and felt less hungry as well (299). However, in all of these studies, a weight loss regimen was essential as well, and without one it is likely that compensation would occur, wiping out any beneficial effects. The 2004 position statement of the American Dietetic Association therefore concluded that, “nonnutritive sweeteners have the potential to promote weight loss in overweight and obese individuals”. Low-calorie sweeteners provide sweetness and palatability to food, with only minimal or virtually no food energy calories. Substituting full-calorie products with low-calorie sweetened product results in fewer calories consumed (244). Assuming that there is no eventual calorie compensation in the regulation of food intake that should be helpful, but such compensation may in fact occur over time without a conscious effort on the dieter’s part to reduce intakes especially when people know they are consuming less calories. Calorie reduction may be achieved only when and as long as low-calorie sweeteners replace their full-calorie counterpart without any additional food eaten to compensate for amount of calories saved.

Because excessive sugar intake might contribute to weight gain and obesity, owing to its effects on increasing caloric intake, a reduction in sugar intake through substitution of low-calorie sweeteners has been proposed to help prevent weight gain. The “AmericanOnTheMove” study in 2007 was conducted to investigate the effect of changes in diet and physical activity on excessive weight gain in overweight children. It showed that a replacement of dietary sugar with a low calorie sweetener (sucralose) in the diet might be an effective tool to reduce caloric intake and reach negative energy balance. However, it should be noted that the experimental group in the study was provided with Splenda products and compensation for participation in the study and these may have been added incentives to patients (300). Thus additional work is needed before the finding can be judged to be well established.

The notion that low calorie sweeteners increase hunger or cravings by uncoupling the sensory and hedonic aspects of sweetness from their satiating effects and thus leading to overconsumption of sugary foods has not been borne out in recent studies. The appetite for sweetness does not seem to be increased by their use and there is no increase in appetite or hunger with consumption of low-calorie sweetened foods and beverages. (301)
(242, 272). One American Dietetic Association’s position statement on nonnutritive sweeteners concluded that nonnutritive sweeteners had no effect on appetite, hunger, and fullness in adults at least, based on results from short-term studies (244). A more recent statement of the Academy of Nutrition and Dietetics in 2012 found that the low calorie sweeteners when substituted for nutritive sweeteners might help consumers to limit carbohydrate and energy intakes as a strategy to manage blood glucose or weight. The American Heart Association/ American Diabetes Association stated in 2012 that some data suggested that low calorie sweeteners might be used in a structured diet to replace sources of added sugars, and that this substitution might result in modest energy intake reductions and weight loss.

Low calorie sweeteners are also claimed to increase adherence in weight-loss programs by providing more palatable food choices from low-calorie sweeteners, and increasing satisfaction with the weight-loss diet (243, 246). These findings also need to be replicated, but it does appear that they have a neutral or slightly positive effect when accompanied by a hypocaloric diet.

Improved quality of the diet

When the caloric intake from added sugar exceeds 25% in usual diets, the amount of some micronutrients in the diet may be reduced and diet quality worsened (244). The theory is that by replacing foods and beverages containing added sugar with the ones containing low-calorie sweeteners, calories can be saved for consumption of more nutrient-dense foods and diet quality may be improved. For example, such products might be helpful for elderly who have decreased energy needs and people who are sedentary and thus require lower caloric intake. However, there is no definitive study to date that explores whether the relationship between use of low-calorie sweeteners and diet quality does in fact exist. One study compared nutrient and energy intakes, quantity of food consumed, and the knowledge and practices between the reduced-sugar foods users and the users of the full sugar versions of the same products (273). The study found that users of reduced-sugar products had higher micronutrient intakes as they reported higher fruits, lower intakes of added fat and sugar, and more label reading. However, this study was cross-sectional and only provided information at one point in time but not over time. Better-designed studies are needed to evaluate relationships between the use of low-calorie products and diet quality.

Diabetes management

Type 2 diabetes is common and appears to be on the rise. The cornerstone of dietary therapy remains weight reduction if the patient is obese, and selected medical options including orlistat (Xenical), phentermine/lopiramate (Quymia) and lorcaserin (Belviq) to assist weight loss and various other medications to assist in the lowering and control of blood glucose levels. Intensive lifestyle interventions that promote weight loss in patients with type 2 diabetes have better outcomes than standard diabetes support and education, including use of fewer medications, lower health care costs and fewer hospitalizations (302). Since low-calorie sweeteners contain virtually no calories and no carbohydrates, they may help people with diabetes to use less sugar and make more healthful food choices, especially if they are trying to lose weight and control their blood sugar levels. However, weight loss will not occur if the dieter substitutes other foods for those that have been eliminated. The American Diabetes Association recommends the use of artificial sweeteners in a calorie controlled diet that also controls carbohydrate intake as part of medical nutrition therapy for diabetes, to better maintain blood glucose levels near normal and control carbohydrate intake (274).

The European Food Safety Authority (EFSA) reviewed data to substantiate health claims related to ability of low-calorie sweetener to reduce post-prandial glycemic response and the use of low-calorie sweetener and glycemic response in people with impaired glucose tolerance. It concluded that such a claim was valid, but that there was no causal relationship between replacement of sugar-sweetened foods and beverages with low-calorie sweeteners and maintenance of normal blood glucose levels (275).
The Glycemic Index (GI) is defined as “the incremental area under the blood glucose response curve of 50 grams carbohydrate portion of a test food expressed as a percentage from a standard food taken by the same subject” (265). Foods with low GI value produce only small rise in blood glucose after ingestion, and thus may aid in control of blood sugar. All sugar alcohols are categorized as low-GI foods. In addition, certain sugar alcohols, sorbitol and xylitol, cause slow increase in plasma glucose due to delayed absorption and metabolism in the liver as well as providing low glycemic responses. Therefore, they may be helpful tools for managing blood glucose levels for people with diabetes. A recent systematic review on Stevia proposed the use of stevia (stevioside) as an antihyperglycemic agent. Stevioside may decrease glucose levels by stimulating the production of insulin, based on studies in humans and animals (262). Another recent study on the effects of stevia, aspartame, and sucrose on postprandial glucose and insulin levels found reduction in postprandial blood glucose levels with consumption of stevia before lunch and dinner meals when compared to both aspartame and sucrose (272). However, better-designed studies on humans are needed to determine the effect of stevioside on glucose tolerance and insulinemia. A recent report hypothesized that low calorie sweeteners increased the risk of altered glucose tolerance and diabetes by altering the microbiome in the gut (Suez et al doi:1036/naturea13794). The claim was based on studies in mice fed very large doses of saccharin, aspartame or sucralose sweeteners which the workers claimed altered insulin resistance. However, when one low calorie sweetener, saccharin, was fed to 7 volunteers, blood sugar rose in 4 and did not change or diminished in the other 3. Other studies with larger sample sizes and more rigorous designs have failed to show adverse effects on glucose tolerance (303)

Reduction of Dental Caries

Dental caries is a complex disease, also involving teeth, bacteria, sugar, time, and saliva. Dietary sugar intake increases the risk of developing dental caries. Sugar (especially retentive forms of sugary foods that stay in the mouth) is metabolized by mutans streptococcus and other cariogenic bacteria on the surfaces of unclean teeth into acids, reducing the pH of the enamel and eroding it, causing decay. When the pH level falls below 5.5, tooth enamel demineralizes through the loss of calcium and phosphate ions (245). Demineralization can be reversed at the stage before the cavity is formed. Decreased sugar intake and good dental hygiene are an important strategy for prevention of dental plaque and dental caries. Therefore, substitution of low-calorie sweeteners (which are not fermented by the bacteria in the mouth) for sugar reduces sugar content of the diet, and thus may help prevent the development of dental caries (276). Sugarless gums and sugarless cough drops, mints, and lozenges also are non-cariogenic, which will not cause dental caries.

It is well known that the low calorie nutritive sweeteners, the sugar alcohols, especially xylitol, have anti-cariogenic properties and prevent formation of dental caries. Polyols are generally not substrates for bacteria in dental plaque. They are not metabolized by the bacteria in dental plaque into acids. The lack of acid production prevents against demineralization of teeth and subsequent dental caries formation (245, 265). However, when polyols (especially sorbitol), are consumed in large amounts (for example, more than two sticks of chewing gum in one day) in animal studies, in one animal study, the number of sorbitol-fermenting bacteria in the mouth increased slightly, somewhat reducing this anti-cariogenic effect. Nevertheless, the fermentation of sorbitol remained very slow when compared to that of sucrose (277). Thus, the bacteria in the mouth adapt to restriction of sugar by alternatively metabolizing sorbitol, but the net effect of the substance still remains positive. More research is needed to on these effects in humans.

Plaque formation may also be affected. In addition to their non-carcinogenicity, polyols also slow demineralization of tooth and promote remineralization of teeth that are with early lesion in the demineralization process. Chewing sugar-free gums containing polyols stimulates more saliva flow in the mouth. Saliva, which has high pH, acts as a buffer to acid production and washes away sugar and acids that could accumulate bacteria. In addition, saliva provides calcium and phosphate ions that can remineralize tooth enamel in the initial stage of demineralization before formation of carious lesion (265, 277).

Among all the polyols, xylitol is the most effective in reducing and preventing dental plaque and caries. Xylitol is the least fermentable polyol. Cariogenic bacteria do not ferment it and thus, the pH of dental plaque does not decrease and the enamel is not eroded (245). Xylitol also inhibits the growth of plaque-forming bacteria themselves (particularly the mutans streptococci) because it is a poor substrate for the bacteria. Therefore, can reduce the accumulation of plaque on tooth enamel (277). The structure of xylitol allows it to form a complex with calcium ions (in a chelate-like structure), which can promote remineralization of tooth enamel, resulting in reversal of the early dental caries lesion (250). Xylitol also acts as a bacteriostatic agent, through the conversion of xylitol into xylitol-5-phosphate by some strains of streptococci. Xylitol-6-phosphate degrades cell membrane of the bacteria and thus, reduces plaque quantity and adhesivity (265). Interestingly, xylitol can also help prevent intra-familial transmission of mutans streptococci from mothers to infants through its property to reduce mutans streptococci quantity (250, 277). Infants can be infected with mutans streptococci through oral transmission from mothers. Infants whose mothers chewed xylitol-sweetened gum had lower counts of mutans streptococci. Therefore, xylitol may also help protect against maternal transmission of cariogenic bacteria.

9.9.3.6 POSSIBLE HEALTH CONCERNS:

Safety

In the USA the Food and Drug Administration permits the sale of all of the low calorie sweeteners mentioned above. In the European Union, cyclamate is also allowed and considered safe.

The products have been judged to be safe for consumption by pregnant women. However, one low calorie sweetener, aspartame (sold as NutraSweet and Equal), should be avoided in pregnancy by women who are homozygous carriers of phenylketonuria because it is metabolized to phenylalanine. In such women at very high levels phenylalanine crosses into fetal circulation and may increase risks of mental retardation. In pregnant women, there is one report that more than one diet drink per day increased risk of preterm delivery (OR 1.38, 1.15-1.65) but this has not been confirmed (304) However, other investigators have not been able to confirm the finding. The European Food Safety Authority recently conducted a full risk assessment on aspartame in view of concerns on the part of consumers that included both animal and human studies. Its’ conclusion, issued in 2013, was that aspartame and its breakdown products were safe for human consumption at current levels of exposure (the ADI for aspartame is 40 mg/kg/day). It added that for patients with phenylketonuria, the ADI was not applicable because they require strict adherence to a diet low in phenylalanine. However threw a no risk to developing fetuses at the current ADI with the exception of women suffering from phenylketonuria. (296). The Dietary Guidelines 2015 Advisory Committee’s draft conclusion for its final report concurred with the EFSA Panel and judged the evidence as moderate that aspartame in amounts commonly consumed was safe and posed minimal health risk for healthy individuals without PKU (including those at risk of most cancers, seizures and cognitive/behavioral problems in children and adults) (305).

Other risks are not well documented. For example, in some observational studies, low calorie sweeteners have been found to be associated with risks of metabolic syndrome (306, 307), with type 2 diabetes (308), or with coronary heart disease and kidney disease (309). However, other observational data show that there is an absence of adverse effects. For example, in the CARDIA study, those consuming a healthy diet pattern with diet beverages showed lower risks of elevated glucose and low HDL cholesterol than did others (310). More impressively in a randomized clinical trial, the CHOICE study, it was found that the diet drinks group showed declines in blood pressure and fasting glucose that were no different than those of controls. The goal was to replace caloric beverages with water or diet beverages to assist weight loss in this randomized single center single blind study of 318 overweight or obese adults studied over a 6 month period. The subject received water alone plus a monthly group website class, a low calories sweetened diet beverage along with the monthly group website, or only the website education on weight loss. None of the groups achieved large weight losses; the largest was the diet beverage group with a loss of 2.5% of body weight, and the smallest was the attention control with 1.5% weight loss at 6 months. The group consuming the diet beverage showed a greater likelihood of achieving a 5% weight loss compared to the attention control (OR 2.29, 95% CI 1.05, 5.01 P=0.04) while the water group did not differ from the control in terms of its likelihood of achieving a 5% weight loss (311, 312). Effects were similar in another study that included weight maintenance. Dieters were given added advice to use the low calorie sweetener aspartame in a yearlong multidisciplinary program of weight loss and two year follow up, while others were not. The differences between the two groups were small but during maintenance the low calorie sweetener users kept more weight off than those who were non-users (267).

Even among those who use many low calorie sweetened foods and beverages, there is little likelihood that they will reach the ADI. For example, one would have to eat 75 low calorie sweetened yoghurts a day for one’s entire lifetime to achieve the ADI for one common low calorie sweetener. In children there is a concern that low calorie sweetened beverages might displace milk and 100% juice, although this fear is not well documented by evidence it has led to regulations that do not include low calorie sweeteners as permitted foods in elementary schools. The American Academy of Pediatrics has recommended that nonnutritive sweeteners should not form a “significant part “ of a child’s diet, and the Academy of Nutrition and Dietetics views them as safe for children within the range of the ADI (244).

Concern has been expressed based on some observational studies, that low calorie sweetened beverages might be associated with long-term weight gain (313, 314), but others show decreased body weight (315), or both increases and decreases (316). However, in a recent met-analysis of 15 randomized controlled trials, which are considered to be superior in assessing causal inference, as well as 9 prospective cohort studies, the correlation between low calorie sweeteners and body weight favored them very clearly over the comparator arms in the randomized studies, and were not significant in the prospective cohort studies (317). The results suggest that other factors, including reverse causation (that is, low calorie sweetener users being more likely to be obese, gaining weight and trying to control their weight gain rather than the sweeteners causing the weight gain may have been present. People who have maintained their weight loss over the long term use fewer sugar sweetened beverages and more low calorie sweetened beverages than to always normal weight persons (318), and in the US National Weight Control Registry long term weight maintainers also used more water, low calorie sweetened beverages and less alcohol and sugary drinks and juices.

Increased appetite and intake

There have been claims of a paradoxical effect of low-calorie sweeteners on appetite stimulation, leading to an increase in food intake. A 1986 study from the United Kingdom (278) compared the effect of water sweetened with aspartame versus plain water on hunger level. Those consuming highly sweetened water rated their hunger level higher than those who consumed only water. However, the study examined only perceived hunger level, which was subjective, and did not really assess participants’ actual food intake. A subsequent study showed no increase in actual food intakes 1 hour after consumption of solutions sweetened with saccharin, aspartame, and acesulfame-K (279).
Another theory of how low-calorie sweeteners might be orexigenic (enhance intake) involves stimulation of food reward pathways in the hypothalamus after ingestion of food (in this case, glucose). Sweetness stimulates the mesolimbic dopaminergic system, producing a feeling of satisfaction and can stimulate food intake (280, 281). Some evidence suggests that such pathway was not observed with ingestion of low-calorie sweeteners. One study using functional magnetic resonance imaging showed longer suppression of this signal in the hypothalamus with glucose ingestion in normal weight men, but found no such effect with sucralose ingestion. Thus, the lack of responsiveness to this signal from consumption of low-calorie sweeteners might theoretically lead to motivation to seek more food (281). However, at present, the evidence is insufficient to conclude that low-calorie sweeteners enhance appetite or food intake, and more research is needed.

Psychobiological signaling between food and the gut as well as energy compensation of meals are two other proposed explanations of how low-calorie sweeteners may lead to overconsumption and weight gain. The psychobiological theory is that low-calorie sweeteners, unlike caloric sweetener, only provide sweetness but not the food energy (282). When sweetness is not accompanied by calories, there is no signal to provoke cephalic phase of digestion in the gut to prepare for arrival of nutrients and to begin the process of energy utilization and thermogenesis. The animal model showed less effective energy regulation through this mechanism, leading to excessive calorie intake and weight gain. In addition, a theory on energy homeostasis has been proposed as another possible mechanism for speculated weight gain from low-calorie sweeteners. Energy compensation, which is the adjustment of energy intake on subsequent meals based on amount of energy consumed on the prior meal, may be disrupted when caloric sweeteners is replaced with low-calorie sweeteners (283). Because low-calorie sweeteners provide only negligible amounts of energy, an upward compensation by increasing intake on subsequent meals is expected. More importantly, some worry that use of low-calorie sweeteners could lead people to believe that they could consume more of other foods to compensate for lower energy from low-calorie sweetened foods and beverages (280). Overall, there is no current evidence on humans to validate any of these theories. However it is clear that inappropriate use of low calorie nutritive or non-nutritive sweeteners that leads those attempting to control their weights to throw all caution in consumption to the winds will nullify any potentially positive effects that they might have.

Low calorie sweeteners have no or modest effects on weight loss and weight maintenance unless accompanied by a hypocaloric diet

A number of studies have been conducted or are now in progress to determine the effects of low-calorie sweetened beverages on
weight changes and metabolic health effects in adults and children in adults, epidemiological studies of children and adolescents are mixed, some showing associations of low calorie sweeteners are associated with increased body weight (319). Two studies (the cross-sectional National Health and Nutrition Examination Survey (NHANES) and the semi-longitudinal San Antonio Heart Study), found positive associations between the use of non-nutritive sweeteners and an increase in body mass index (BMI) (268). However, the design was such that it is not clear if obese people used more low-calorie sweeteners (more likely) or whether low-calorie sweeteners caused them to be obese (less likely). Another longitudinal cohort study found a positive correlation between increased consumption of diet soda and BMI z-scores after two years in 164 elementary school aged children (284). However, the study design again was not such that it could demonstrate a causal relationship between the two, possibly the effects were again due to reverse confounding (e.g. the obese who are most likely to gain weight were those who also used more diet products). Attributes of causal relationships such as strength of association, temporal relationship, consistency of findings, biological plausibility, and strong dose-response relationship, need to be considered when epidemiologic studies of low-calorie sweetener consumption and weight gain are examined. In the few small randomized trials of overweight and obese children, low calorie sweeteners do not or only minimally decrease weight and body mass index (320, 321, 322). Therefore, although there is no strong evidence that use of low-calorie non-nutritive sweeteners (especially in beverages) increases weight, there is also little evidence that it decreases it substantially. However, adding several interventions, such as was done in the 6 month America on the Move study, in which 192 families with at least 1 overweight child were randomized to either an intervention involving increasing physical activity by 2000 steps a day and decreasing sugar by 100 Calories per day with a low calorie sweetener or to a control group which involved only self-monitoring, the two interventions together did show small but greater changes in body mass index for age among the children at month 6 (323).
Another theory to explain the purported association between low-calories sweeteners and weight gain is that lower carbohydrate intake may result in higher fat intake. A reduction in carbohydrate intake by replacing sucrose with low-calorie sweetener is claimed to lead to a higher proportion of energy coming from fat, which in turn may lead to weight gain. However, accumulating evidence does not support the claim that macronutrient shifts occur from the replacement of low-calorie sweeteners and an increased fat intake in the diet (280).
Another potential, but unproven theory based on a study in rats linking low-calorie sweeteners and weight gain is that the gut microflora are altered when they are exposed to low-calories sweeteners. Changes in gut microflora might then trigger inflammatory pathways; promoting insulin resistance, fat accumulation, and weight gain in the individual (286). There is no evidence in humans to support this claim, and the evidence in experimental animals is mixed at best..

Development of a “sweet tooth” due to low calorie sweeteners?

Artificially sweetened and calorically sweetened foods and beverages are claimed to increase the preference for a sweet taste, with an increase in consumption of artificial sweetened beverages may change taste preferences toward sweet foods, especially in children (268). This preference is feared by some to bring about the replacement of healthful foods with sweets, and thus may be associated with lower diet quality in children. However, at present little evidence exists that this is so. In addition, repeated exposure to sweetness could lead to an increased acceptance of sweet sensation as a result of learned behavior and the exposure to sweetness in low-calorie sweetener might establish preference to sweetness in the same manner as other sugars (280).

Low-calorie non-nutritive sweeteners have been shown to activate sweet-taste receptors in the gut in the same manner as sugar does in animal studies. T1R2 and T1R3 are transmembrane sweet-taste receptor proteins found in the gut of both rodents and humans (286, 287). Compounds with a sweet-taste (e.g. sugar and low-calorie sweeteners) bind to these receptors, stimulating signaling pathway to the brain in experimental animals. This leads to changes in electrophysiological patterns in the brain, resulting in preference of sweetness. At present, the evidence that low-calorie sweeteners encourage or exacerbate sweet tooth remains scattered. More research is needed.

What can be said with certainty is that infants and young children do not require low-calorie sweeteners although they are judged as safe by the FDA. The American Dietetic Association (ADA) suggests that parents of children less than 2 years of age discuss use of those products with their pediatricians to ensure that childrens’ needs are met (244). Concerns about ensuring that children and adolescents learn good food habits have led some individuals and expert groups to recommend that schools restrict the use of calorically sweetened beverages and high-calorie, low nutrient density sweet foods that are also high in added sugars (288). However, some of those reports have also recommended against the sale of low-calorie sweetened beverages in school, not for safety reasons, but to foster nutrition education. The rationale for doing so is not entirely clear, but nonetheless, many elementary and secondary schools now do not permit non-nutritive sweetened or calorically sweetened beverages in schools. Research is needed to determine if such a prohibition has intended effects on nutrition education, development of a “sweet tooth”, dental caries, weight gain or maintenance, overall intakes of sugar, added sugar, or food energy, or other objective standpoints. Also, unintended effects, such as more extensive overall use of the products as gestures of defiance must be assessed.

Metabolic Syndrome

Some large prospective cohort studies found associations between intakes of low-calorie sweeteners and incidence of metabolic syndrome, but the designs were such that cause and effect could not be demonstrated (289). Proponents that such a link is causal suggest that the association is due to deregulation of glucose homeostasis caused by the use of artificial sweeteners. Another, simpler, explanation is a reverse causation. One study on consumption of diet soda in young healthy volunteers before an oral glucose challenge showed an increase in secretion of GLP-1, the anti-hyperglycemic hormone released by intestinal cells, which can alter gastric emptying and secretion of insulin. Whether the low-calorie sweeteners in fact had these effects is not yet clear; the study has not been replicated. The concern raised is that consumption of artificial sweeteners together with foods and drinks containing sugar may lead to increased GLP-1 and insulin secretion, which may result in more rapid sugar absorption and perhaps in turn, could influence blood sugar levels. However, at present, evidence is lacking that this is the case.

Evidence from in vitro and rat studies suggests that low-calorie sweeteners stimulate the sweet-taste receptors in the gut, causing the up-regulation of transporters, sodium-dependent glucose transporter (SGLT 1) and glucose transporter 2 (GLUT 2), on the apical membrane of the small intestine to increase glucose uptake. In addition, low-calorie sweeteners (especially sucralose and saccharin) may also stimulate release of incretin (GLP-1), a gut hormone that stimulates insulin release after ingestion of glucose. This cascade of intestinal uptake transporters, incretin, and insulin release may disrupt glucose homeostasis (286, 287). However, extensive number of studies conducted in humans showed no such effect. Overall, there is no consistent evidence in humans that low-calorie non-nutritive or nutritive sweeteners have adverse effects on insulin release and blood glucose homeostasis.

9.9.3.7 SUMMARY AND RECOMMENDATIONS

  • None of the low-calorie sweeteners permitted by FDA on the US market are necessary in human diets or for human health. However, some consumers enjoy the sweet taste of nutritive or non-nutritive low calorie sweeteners and there is no reason to discourage their use on safety or health grounds.
  • All FDA-approved sweeteners are safe for use by the general population, pregnant women (except phenylketonurics), and children (except phenylketonurics) as doses below the Acceptable Daily Intake (ADI). For pregnant women (other than those with PKU) the American Academy of Pediatrics has stated that aspartame is safe for pregnant women and their developing infants. There is also evidence that acesulfame K, sucralose, and the sugar alcohols are safe for pregnant women in small amounts. Little research has been conducted on the safety of saccharin and stevia in pregnant women.
  • None of the low-calorie nutritive or non-nutritive sweeteners are perfect substitutes for sugar from the standpoint of taste, mouth feel, and functionality in food processing and preparation, but they are acceptable to many people.
  • Most of the nutritive and non-nutritive low-calorie sweeteners are non-cariogenic. Xylitol also appears to have an anti-cariogenic property as well, enhancing remineralization of tooth enamel.
  • The best-documented adverse effects with low-calorie sweeteners are from poorly absorbed nutritive low-calorie sweeteners (sugar alcohols) that have laxative effects when consumed in large amounts. A theoretical hazard exists for aspartame with very heavy use of individuals with phenylketonuria (PKU). Other health concerns, such as the fostering of a “sweet tooth” that leads to excessive energy intakes are poorly documented.
  • Low calorie sweeteners may be helpful in weight control if they are substituted for some of the caloric sweeteners in the diet and calorie intakes are also reduced. Otherwise, they appear to have little effect. Their use alone without healthful eating, physical activity, and behavior change is unlikely to be helpful in weight management.
  • Low-calorie sweeteners contribute little or nothing to the glycemic response and as such may have advantages over caloric sweeteners for individuals who need to control their blood sugar levels.
  • There is no current scientific reason to recommend against the use of low calorie sweeteners for those who are trying to lose weight and wish to use them.
  • For patients or other users who are concerned about excessive intakes of nutritive or non-nutritive low calorie sweeteners, the best advice is to use several different types of low-calorie sweeteners so that doses of any one compound are low and to always stay below the ADI, or to not use them.

Table 20 is a description of nutritive and nonnutritive low-calorie sweeteners approved by the FDA or recognized as generally recognized as Safe (GRAS). The table describes the following for each non-nutritive and nutritive sweetener; brand name, definition, characteristics, metabolism and excretion, chemical structures, sweetness, calories, ADI, year approved, uses, health benefits, health concerns and additional comments.

Table 20. Nutritive and Non-nutritive low-calorie sweeteners approved by FDA or recognized as Generally Recognized as Safe (GRAS) (242-291).
Non-nutritive sweeteners Nutritive Sweeteners
Names Aspartame Acesulfame-K Saccharin Sucralose Neotame Advantame Steviosides Mannitol Xylitol Sorbitol Erythritol
Brand names NutraSweet®, Equal®, others Sunett®, Sweet One® Sweet’N Low®, Sweet Twin, Sugar Twin®, Necta Sweet® Splenda® Used as ingredient in food products. Used as an ingredient in food and beverage products Stevia®, Truvia™, Sun Crystals®, PureVia™, Sweetleaf Sweetener™ Used as ingredient in food products. XyloSweet Used as ingredient in food products. Zerose
Definition Synthetic sweetener composed of aspartic acid and phenylalanine. A combination of an organic acid and potassium. Synthetic sweetener in forms of sodium or calcium saccharin. A sugar derivative by replacing 3 hydroxyl groups with 3 chlorine atoms on the sugar molecule. Dipeptide methyl ester derived from aspartic acids and phenylalanine. Synthetic sweetener produced in a 3-step process that ultimately combines aspartame and HMPA Derived from the leaves of Stevia rebaudiana plant in South America. Known as “sweet leaf.” A hexose alcohol extracted from seaweed. An intermediate product of carbohydrate metabolism from xylan-containing plants. A hexose alcohol from hydrogenation of glucose and fructose with nickel catalyst. A tetrose alcohol derived from the cultivation of yeast-like fungi on glucose.
Characteristics Loses sweetness with high heat. Highly heat stable for cooking and baking. Metallic aftertaste. Highly heat stable for cooking and baking. Bitter metallic aftertaste. Highly heat stable for cooking and baking. Highly heat stable for cooking and baking. Clean sweet sucrose-like taste. Heat Stable for cooking and baking. Clean sweet sucrose like taste. Ultra high potency. Heat stable. Licorice aftertaste. Enhances sweet and savory flavors. Lacks bulking property. Heat stable. High melting point. Non-hygroscopic(does not pick up moisture). Sweetest of sugar alcohols. Quickly dissolves. Produces cooling effect in the mouth. Heat stable and highly soluble. Does not cause browning. Humectant (retain moisture). Very water-soluble. Non-hygroscopic.
Non-nutritive sweeteners Nutritive Sweeteners
Metabolism and Excretion Broken down into aspartic acid, phenylalanine, and methanol upon digestion. All compounds are metabolized normally, except in individuals with PKU. Not metabolized and excreted unchanged by kidneys. Not metabolized and excreted unchanged by kidneys. Not randomized and excreted by the kidneys and in feces. Partially absorbed and excreted in feces and urine.   Not absorbed in small intestine. Degraded into steviol by bacteria in the colon, where it is absorbed. Excreted in the feces and urine. 25% is absorbed and excreted in the urine. Unabsorbed portion is fermented by colonic bacteria. 50% absorbed and excreted. Unabsorbed portion is fermented by colonic bacteria. 25% is absorbed and excreted in the urine. Unabsorbed portion is fermented by colonic bacteria. 90% is absorbed. Rapidly excreted in the urine and feces within 24 hours.
Relative sweetness compared to sucrose* 180 200 300 600 7000 - 13000 20000 200 - 300 0.5 - 0.7 1 0.5 - 0.7 0.6 - 0.8
Kcal/g 4 0 0 0 0 0 0 1.6 2,4 2,6 0.2
ADI (mg/kg/d) ** 50 15 5 5 18mg / NA 1970 mg/day 0-4 (as steviol) Not specified. Not specified. Not specified. Not specified.
ADI for 70kg person / Cans of soda equivalent 3500mg / 28 1050mg / 21 350mg / 4 350mg / 6 18mg / NA 1970 mg/ NA 0 – 280mg / 5 NA / NA NA / NA NA / NA NA / NA
Year of approval by FDA and as GRAS. 1981 1988 Prior to 1958. Reapproved again in 2000. 1998 2002 2014 GRAS in 2008 1986 1983 GRAS 1982 GRAS in 2001
Chemical Structures Aspartame Acesulfa me-K Saccharin Sucralose Neotame Advantame Steviosides Mannitol Xylitol Sorbitol Erythrit
Non-nutritive sweeteners Nutritive Sweeteners
Uses Tabletop sweetener, ingredients in foods and diet soft drinks. Limited use in bakery products. Tabletop sweeteners, baked goods, frozen desserts, candies, beverages, cough drops, and breath mints. Tabletop sweetener, soft drinks, baked goods, jams, chewing gum, canned fruit, candy, dessert toppings, salad dressings. Tabletop sweetener, beverages, chewing gum, frozen desserts, fruit juices, gelatins. Flavor enhancer, baked goods, soft drinks, chewing gum, frozen desserts, jams, puddings, gelatins, processed fruits. Flavor enhancer, baked goods, soft drinks, chewing gum, frozen desserts, jams, puddings, gelatins, processed fruits. Tabletop sweetener, juices, tea beverages. (Used extensively in Japan for pickles, dried seafoods, and confections). Dusting powder for chewing gum, ingredient in chocolate-flavored coating agents for ice cream and confections. Chewing gum, hard candy, oral health products, cough syrups and cough drops, children’s chewable multivitamins, foods for special dietary needs. Sugar-free candies, chewing gums, frozen desserts, pastries Bulk sweetener in diet food products, candies, beverages, fat-based creams, chewing gums, confection, yogurt.
Health benefits Virtually calorie free. Calorie free. Calorie free. Calorie free. Calorie free. Calorie free. Calorie free. Claimed to have a hypoglycemic effect. Low calorie content. Non-cariogenic. Low glycemic response. Low calorie content. Reduces dental plaque and caries and may promote tooth remineralization. Low glycemic response. Low calorie content. Slow absorption and metabolism independently of insulin might benefit for diabetics. Calorie free.
Unlikely to have a laxative effect. Non-cariogenic.. Low glycemic response.
Non-nutritive sweeteners Nutritive Sweeteners
Health concerns   All should be used at levels below the ADI. Strong laxative effect at >20 mg/day. Strong laxative (> 50 mg/day) and also diuretic effects. Flatulence and diarrhea.  
Comment Requires a label that product contains phenylalanine.       Does not require a label for phenylalanine content due to negligible amount used and low availability of phenylalanine from the neotame.     Requires a warning label for a possible laxative effect.   Requires a warning label for a possible laxative effect.  
*Relative sweetness as compared to sucrose (table sugar). 1= reference value which is the sweetness of sucrose.
** ADI = Acceptable Daily Intake
*** Other non-nutritive low-calorie sweeteners (Alitame, Thaumatin, Neohesteridine, and Glycyrrhizin) are not yet approved as both sweeteners and as GRAS in the US. See text for details.http://beverageinstitute.org/

9.10 Energy Density

Energy density, or caloric density, is defined as the calories provided per unit weight of food eaten (such as calories/gram). When the composition of a diet of usual foods is decreased in fat, the energy density of the diet tends to fall since the total weight of food consumed remains constant, or may increase (196-198) . Some of the beneficial effects of low fat diets in weight loss and maintenance may be due, at least in part, to low energy-density, which may promote satiety. The premise is that if foods bring more weight than calories into the body, gastric distention and intestinal bulking may promote fullness. For example, foods high in water and/or fiber tend to have low energy density, with some that are very high in volume, such as unbuttered popcorn. Their inclusion in a weight reduction diet is advocated by some experts for this reason (10), and because they might increase overall dietary quality (199). As will be discussed later, some evidence suggests that low-energy density, high-volume diets may help people ingest fewer calories and thus may assist with weight loss, although longer-term research is needed (200; 201;199).

9.11 Volume of Food

Low energy density is the basis of the “Volumetrics” diet put forth by Dr. Barbara Rolls. This diet is high in fruits, vegetables, cooked whole grains and fibrous foods, which are high in water and bulk, to add volume for satiety without extra calories (285). In a one year study completed by 71 obese women, the low energy density diet group showed slightly better weight loss than a low fat diet group, and reported lower hunger (324). Cross-sectional work has shown that people who maintain large
amounts of weight loss over years tend to eat a lower energy density diet than normal weight or obese subjects (201). Some other diets tend to be low in energy density without specifically targeting energy density, through their inclusion of fruits, vegetables, and fibrous foods (199).

10 Available Programs

According to the U.S. Food and Drug Administration (FDA), Americans spent an estimated $30 billion in 1992 on diet and weight loss programs. Market data, an independent market research and consulting firm, has estimated annual spending on diet and weight loss programs to have reached $61 billion by 2013 (203). The number of products and programs available to consumers is essentially endless, and the quality of what is available can be questionable. Therefore, patients should be advised to research programs or products they are interested in, and consult with a physician before utilizing any commercial weight loss option.

10.1 Registered Dietitians: Dietetic Advice and Individualized Eating Plans

A registered dietitian is a food and nutrition expert who has met academic and professional requirements including:

  • A bachelor’s degree at an accredited university, approved by the Commission on Accreditation for Dietetics Education (CADE) of the American Dietetic Association (ADA).
  • Completed an accredited, supervised, experiential practice program (dietetic internship) at a health-care facility, community agency, and/or foodservice corporation, including at least 900 hours of hands-on experience.
  • Pass a national examination administered by the Commission on Dietetic Registration (CDR).
  • Complete continuing professional education requirements to maintain registration.

Many physicians lack the time that obese patients require for successful weight control therapy. Referral for dietary counseling to a registered dietitian (RD) is useful for many patients, particularly those who have comorbidities that also require medical nutrition therapy. Many registered dietitians also hold certifications in specialized areas of practice, including weight management, providing additional expertise in the management and treatment of obesity (203). They are able to understand more complicated therapeutic dietary recommendations that many patients, especially those with comorbidities, require. Practicing registered dietitians in your area can be located using the website for the Academy of Nutrition and Dietetics at http://www.eatright.org/.

 

10.1.1 Available Programs

There are over 67,000 registered dietitians in the United States, practicing in hospitals, outpatient centers, health centers, the community, and in private practices, among many other areas of expertise (204). Patients can see a registered dietitian via patient referrals from physicians for a variety of health problems that require dietary modification, or by self-made appointments. Depending on the area of dietary modification/treatment, health insurance may, or may not, cover the services provided, so patients need to check with their insurance providers before scheduling an appointment. Some formal weight control programs staffed by dietitians are available in hospitals and health centers, where individual counseling is also available.

10.1.2 Candidates for Care

Dietary advice of a general nature is not enough for patients who have multiple comorbidities requiring medical nutrition therapy (e.g., diabetes, hypertension, coronary artery disease, gastrointestinal disorders, etc.), those on multiple medications, and those with complex and involved health problems that have dietary implications. These patients are prime candidates for dietetic therapy with a dietitian. Patients who have had poor outcomes in weight control efforts on their own, who have special dietary needs or preferences, and who need extensive education and assistance are also particularly likely to benefit.

10.1.3 Advantages

Registered dietitians are able to read and interpret medical records and are equipped to adopt weight loss prescriptions to the particular needs of patients. Their knowledge of food habits, food preparation, and food products on the market makes them an excellent resource for helping patients to adopt the general weight control prescription to ta patient’s particular circumstances. A particular advantage of dietetic involvement in patient care is that dietitians often work in medical settings and have access to patient charts, as well as the ability to consult with other health professionals. Registered dietitians are helpful in treating patients on multiple medications, on very-low-calorie diets, and post-gastric bypass counseling. Some dietitians have advanced certification in weight management, and are especially well equipped to counsel patients with complex and involved medical problems. Some insurance companies and health maintenance organizations may pay for obesity treatment when it is part of a larger therapeutic program for conditions such as diabetes if it involves a dietitian who is a certified Medicare provider. Patients should check with their insurance providers about reimbursement.

10.1.4 Disadvantages

The patient’s out-of-pocket costs for dietary counseling from a registered dietitian vary, depending on insurance coverage and the comorbidities that need to be treated/addressed. It is important to note that personal trainers and/or nutritionists do not necessarily have the credentials to be counseling patients on nutrition and dieting. This is particularly true of patients who have comorbidities. . Providers should encourage patients seeking counseling on their own to look for the “registered dietitian” credential.

10.1.5 Safety and Effectiveness of Therapy

Registered dietitians are health professionals who go through extensive schooling and training and are registered in a national registry with specific standards. Licensure is also required in 31 states. Registered dietitians, therefore, have medical, legal, and ethical obligations to their patients. Their own education includes formal educational requirements of at least a baccalaureate degree, a dietetic internship, supervised clinical training, a registration exam, and mandatory continuing education. Dietitians are trained to read medical charts, to work with physicians and other allied health professionals, and to alert physicians when untoward events arise. Thus, their recommendations regarding weight loss are likely to be safe and evidenced-based. The effectiveness of dietetic counseling, like that of physician counseling for weight control, has seldom been evaluated.

10.2 Commercial Weight Loss Programs

Commercial non-medical weight control programs are popular and widely available in the United States and Canada. They will be discussed in detail in the following section.

10.2.1 Available Programs

Commercial programs include large chains such as Weight Watchers®, Jenny Craig®, LA Weight Loss Centers®, Nutrisystem®, and many regional ventures. These programs vary, but generally include advice on a structured low calorie diet, exercise, lifestyle modification coupled with group support and/or individual counseling. Oftentimes, there are options for delivery of pre-portioned reduced-calorie meals. Usually the program is administered by a layperson trained by the program who is often a successful program graduate. However, laypersons trained by the company and degree-trained professionals (such as dietitians) may also be on their staff. It should be noted that these programs do not provide physician supervision although they usually require physician sign-off before involvement (105). All of these programs are for-profit entities and charge fees (196). With the growth of the Internet, many programs now offer online support as an adjunct or replacement to more traditional in-person individual or group counseling. A sample of some popular commercial programs is outlined below in Table 21.

Table 21. Popular Commercial Programs
Product Comments
Jenny Craig®
(Nestle Nutrition®)
www.jennycraig.com
A commercial program where dieters are paired with trained consultants, often program graduates, who help set goals, plan weekly meal plans, and provide support through in person and phone meetings. The program also includes home-delivered pre-portioned food that provided 3 meals and a dessert or snack each day. The meals are designed to promote 1-2 lbs of weight loss each week using portion control. The plan starts at $19 per month, plus the cost of food, which ranges from $16-$23 per day. A full line of products is also available to be purchased outside the official program in retail outlets. Options in this line range from Complete Meals to Café Steamers. Meals are between 180-410 calories and are available at most local grocers. There are also desert options. These products fulfill American Heart Association heart checklist program criteria, and are lower in sodium than some other brands
Nutrisystem®
(Nutrisystem Inc. ®)
www.nutrisytem.com
Participants in the program receive all meals, snacks, and desserts via home delivery. Foods may be chosen a la carte or according to a meal plan with predetermined food choices. Various plans are offered for men and women in different categories: basic for the budget conscious, core for added convenience and variety, select for the most variety, diabetic for diabetics, and vegetarian for vegetarians. Prices vary depending on the plan, but are expensive. One month on the program can cost between $300 and $500, and may be more expensive. The program also includes access to an online community and phone counseling. 5-day weight loss kits are also available in retailers such as Wal-Mart® and Costco® for around $50.00.
Weight Watchers®
www.weightwatchers.com
Weight Watchers uses a group support focus to promote weight loss through attendance at weekly meetings, where members support each other, discuss challenges and successes, and weigh-in. Weight loss is achieved by teaching dieters how to subscribe points to a variety of different foods and eat within a certain point budget each day. For around $10 per week, dieters can attend meetings and have access to online tools and a mobile app. There is also an online-only self-help version of the Weight Watchers® program which provides a diet plan and fitness information with exercises. Cost is currently around $20 per month, with an approximately $30 sign-up fee. However, the often run cost-saving promotions for those willing to sign-up for at least 3 months.
LA Weight Loss Centers®
www.laweightloss.com
A 3-part plan that provides meal plans with recipes, snack bars, and dietary supplements. Other tools available include educational material, food diaries, and specialized plates and containers to aid in portion control. The silver plan cost between $150-$200 per month, and does not include supplements, while the gold plan costs between $200-$250 per month and offers supplements designed by the company called “nutritionals”. Support from counselors is available in-center and online.
Ediets.com
www.ediets.com
Provides a personalized reducing diet and food list, fitness information, healthy recipes, social networking community, and charts and dieting tools. There are 3 categories of diet plan currently available: the “Vitabot plan”, the “Holly Madison Diet,” and the “Nutrihand plan.” Cost is currently $9.95 per month. Meal delivery options are also available for $30 - $40 per day through “The Chef’s Diet,” which uses a 40-30-30 ratio of carbohydrate, protein, and fat, respectively, and between 1300-1500 calories per day. This option includes 3 meals per day and 2 snacks
Diets.comwww.diets.com A nutrition and health website with tools to aid in weight loss, healthy living, and wellness. Basic membership is free and includes access to online support groups, articles, and useful tracking tools. However, premium membership, including all of the above in conjunction with a customized diet plan, personalized exercise plan and coping plan, personalized “expert” advice, and individualized weekly self-checklists to help keep dieters on track, is available for around $40 per month, with those willing to sign-up for 6 months getting the largest discount at just over $14 per month.

10.2.2 Candidates

Overweight and moderately obese persons with few risk factors and few comorbidities are good candidates for these programs. Those who find that they need continued motivation, monitoring, and social support with a structured regimen may particularly benefit from one of these programs.

10.2.3 Appropriate Use

These programs are not substitutes for physician concern for or medical monitoring of his or her patients’ weights. They are most successful when the patient’s personal physician continues to provide encouragement and supervision because most commercial weight loss programs provide no or very little physician supervision. The commercial programs are not equipped to deal with patients with multiple involved comorbidities of either a medical or psychological nature. Patients with complex medical issues are better treated by a program and therapists who are more closely connected to the health care system where medical charts and other patient-specific information is available. Registered dietitians and specialized weight control programs operated by medical facilities are more appropriate options in this population.

10.2.4 Advantages

Most major established commercial chains offer well-crafted, nutritionally adequate, and behaviorally sound programs that, overall, are reasonable therapies. Classes are often held in places of employment or neighborhood centers that are conveniently located. Weight Watchers® offers frozen entrées and other weight control products that are integrated into the program and available in supermarkets, making adherence easier. Jenny Craig® and LA Weight Loss Centers® also offer frozen entrees and various weight control products; however, these are only available through their stores. On the Nutrisystem® program, the customer must eat only Nutrisystem® food for a defined period based on individual needs. Nutrisystem® sends all food items including snacks to the customer via mail.
In a multicenter, randomized, two-year study of 423 subjects with a BMI of 27 to 40 kilogram/m2 it was shown that a structured commercial weight-loss program was more likely to be effective for managing moderately overweight patients than brief counseling and self-help (205). Individuals were randomly assigned to either a self-help program, consisting of two 20-minute sessions with a nutritionist and provision of printed materials and other self-help resources, or to attendance at meetings of a commercial program (Weight Watchers®). After 26 weeks subjects in the commercial weight-loss program had greater decreases in body weight, BMI, mean waist circumference, and fat mass (205). It is important to discuss commercial program options with individuals so they know their options, but only with individuals who are plausible candidates.
Since it was founded in 1997, most of the large commercial programs have joined the Partnership for Healthy Weight Management, a voluntary association. Members provide, on a voluntary basis, publicly available information to help potential participants meet their needs. Criteria for membership require that programs disclose staff qualifications, essential components of the program, the risks associated with overweight and obesity, other details about the provider’s program or product, and program costs.

 

10.2.5 Disadvantages

Because they are profit-driven business, the main objective of a commercial weight loss programs may not be driven by patient care. Although most programs require physician approval before participants can enroll, there is no guarantee of the quality of the health assessment that has been carried out prior to enrollment. For some individuals, especially those at very high risk, more intensive medical supervision may be required. The cost of the programs is another obstacle. Many of the poor who are obese do not have the resources to purchase these services and products, even though they might benefit from them. Discounts or waivers of fees for those in financial hardship are rarely available.
Statistics are rarely kept on success rates or long-term adherence. Another problem is maintenance of lost weight and preventing relapse. The companies have become more active in developing programs catering to those who have lost weight to help them maintain their losses in recent years, but incentives to patients for staying in maintenance programs may still not be sufficient. Additionally, very few high-quality studies have assessed the efficacy of commercial weight loss programs and the ones that do provide the best-case scenario for results—as they do not account for participants who have dropped out of the program (206). The only program that has published high-quality studies to date is Weight Watchers®. The best study on Weight Watchers® determined that participants lost 5% of their initial body weight (about 10 pounds) in 6 months and kept off 3% (about five pounds) at two years (206).

 

10.2.6 Effectiveness and Safety

The major firms provide programs and products that are safe for patients without major comorbidities when directions are followed. However, in spite of the fact that millions of Americans have purchased these services, their effectiveness in bringing about weight loss or sustaining lower weights has rarely been studied with scientific rigor (105).

10.3 Formulas and Meal Replacements

In addition to commercial weight loss programs, many meal replacement and formula products for weight control are now available. Patients can purchase these products on their own in supermarkets, drug stores, and online. Unlike very low-calorie diet formulas, which are medical foods that are usually provided as part of a medically supervised treatment program (see Table 13), these products can be purchased by anyone.

10.3.1 Available Products

Meal replacements now include not only powders like Slim-Fast® that are mixed with milk or other liquids, but drinks, bars, and frozen entrees. Formulations and nutrient content vary. Most liquid meal replacement products provide about 220 calories per serving and are relatively high in protein, vitamins and minerals, but low in fat (see Table 22 for examples of over-the-counter, ready-to-drink, liquid meal replacements). The health bars and frozen entrees vary in their caloric content, but are generally between 200 and 400 calories, and have more complete profile of nutrients. The entrees include offerings such as Lean Cuisine®, Healthy Choice®, and Smart Ones®, among others (see Table 23 for product listing). All of these pre-packaged entrees share characteristics such as discrete portion sizes that are relatively low in calories (usually 300 calories or less). Generally, all frozen entrees are high in sodium, with at least 500 mg of sodium per serving. Smart Ones® is manufactured by HJ Heinz, and is closely allied with the Weight Watchers® commercial diet program. Its packages are prepared to fit into the food plans for the Weight Watchers® program. All of the meal replacement products are designed to be eaten with additions of conventional foods that supply dietary fiber, other nutrients, additional calories and fluids.

Table 22. Examples of Over the Counter, Ready to Drink, Liquid Meal Replacements for Weight Loss
  Total calories Size % Carbohydrate %Protein %Fat
GNC® Total LeanTM Lean Shake TM
(GNC®)
170 14 fl oz 6 25 6
EAS® Myoplex® Lite
(Abbott®)
170 11 fl oz 20 20 2
Slim-Fast® Protein Meal Shakes
(Unilever®)
180 10 fl oz 4 20 9
Atkins™ Advantage® Shakes
(Atkins Nutritionals®)
160 11 fl oz 6 15 9
Glucerna® Hunger SmartTM Shake
(Abbott®)
180 11.5 fl oz 16 15 8
Carnation® Breakfast Essentials™ No Sugar Added Complete Nutritional Drink (Ready-to-Drink)
(Nestle Nutrition®)
250 11 fl oz 16 13 5
Note – Glucerna is formulated for individuals with diabetes or prediabetes
Table 23. Popular Frozen Entrees
Product Comments
Healthy Choice®(ConAgra Foods®) A full line of products from Complete Meals to Café Steamers. Meals are between 180-410 calories and are available at most local grocers. There are also desert options. These products fulfill American Heart Association heart checklist program criteria and are lower in sodium than some other brands
Kashi® Frozen Entrées(Kashi®) A variety of different entrée options are available, many of which are vegetarian. Typically, these entrees range from 250 to 400 calories. They are available in local grocery stores.
Lean Cuisine®(Nestle Nutrition ®) A wide range of entrees from Panini sandwiches to lasagna. Entrées, including breakfast options, are between 140-400 calories, and are available at many grocery stores.
Smart Ones®(Heinz®) Smart Ones® products are associated with the Weight Watchers® program, and available at many grocery stores. Entrees range from 180 to 310 calories, and packaging also includes the Weight Watchers® points associated with each item. A small tossed salad and/or fruit may be added to make the meal more complete. There are also snack, desert, and breakfast options available.

10.3.2 Candidates

Individuals who are healthy but moderately overweight (BMI 25-30) and who wish to lose less than 5% of their body weight or who wish to use these products for one meal a day to assist in their weight maintenance efforts may find these products helpful. The products provide an easily prepared, generally nutritious, and relatively modest caloric load that can satisfy hunger. For those who are susceptible to environmental triggers (such as being involved in meal preparation or eating in cafeterias or fast food restaurants) and respond by overeating, these products offer a safe and palatable option that lessens temptation.

10.3.3 Appropriate Use

Portion controlled liquid meal replacements such as Slim-Fast® or Shakeology (and many other products) are recommended for two meals and a snack with a small meal of conventional foods and low or no calorie beverages. They should not be used as the sole source of nourishment on a diet. The entrée choices are suitable for meals, but their use for multiple meals a day should be cautioned in patients for whom sodium consumption is a concern.

10.3.4 Advantages

The main advantages of meal replacements are built-in portion and calorie control, widespread availability, convenience composition that is fairly micronutrient dense while remaining low in calories, ease of preparation, and for some of the dry or canned products, portability. Costs of the meal replacements are reasonable, and can simplify food choice decisions. They are lower in calories than many snack or restaurant foods that people who are eating away from home might otherwise consume. They are also convenient, rapidly and easily prepared, and can be eaten anywhere, allowing eaters to avoid “high risk” eating environments.

10.3.5 Disadvantages

The major disadvantages of these products are their cost, monotony, and limited variety. From a nutritional standpoint, the products vary, but are often quite high in sodium (600 plus milligrams per serving). Only Healthy Choice® is low in calories, saturated fat, and also in sodium. As with most strategies, they are ineffective unless they are used as part of an overall low calorie eating plan. If they are used as sole sources of food they would be nutritionally inadequate not only in energy, but several other nutrients and water. Additionally, they might not provide a patient trying to lose weight with practice in planning and preparing their own healthy low-calorie meals for a lifetime of healthy weight management. However, there is little evidence on this part.

10.3.6 Effectiveness and Safety

These products may be nutritionally inadequate when they are used as the sole sources of food and fluids for many weeks. When the products are used according to directions on the label or in package inserts, they are safe (71) . When used as part of a weight loss program these single meal replacements are effective during the weight loss phase (198). They are also valuable additions in the weight maintenance phase, often because the meal replacements provide a low set number of calories in an easy-to-fix-entrée (5), with control over portion size (207).

10.4 Weight Loss Books and Manuals

In addition to weight loss products, Table 24 provides some examples of popular diet books. Books are difficult to use on one’s own because there is little reinforcement. The quality of self-help books on weight control ranges from the sublime to the ridiculous. Among the better, older books currently on the market are the LEARN® Program for Weight Management, which is a sound 15 week course that is usually administered within a treatment program (208). The book is effective when it is part of the treatment program. However, the charges for such a program are considerable, the program is not available in all parts of the country, the effectiveness of self-directed efforts using the book by itself has not been evaluated, and the dietary advice is often vague. Another good book is Volumetrics by Barbara Rolls PhD (209), which encourages a diet based on foods that have a low energy density, meaning that they contain few calories per gram of weight. Dr. Rolls’ research has shown that foods with large volume but few calories can provide satiety while helping individuals avoid over-consumption of energy. Such foods are usually high in water and fiber, while low in fat. Although the long-term efficacy of this specific diet has yet to be affirmed, the diet is rich in fruits, vegetables, and other healthful foods (209). The bottom line on diet books is that with few exceptions, the dieter is sure to lose his or her money , but whether weight is lost or not is less certain. Moreover, in spite of the hype there is little evidence that, aside from the few books mentioned above, that the diets “work” and that the authors have discovered a unique new scientific principle that causes weight loss.

 

Table 21. Popular Diet Programs and Books (46)
Diet Brief description Average Calories Per Day Composition % of Calories Type of Diet
      %CHO %Protein %Fat  
5-Factor DietHarley PasternakBallantine Books, 2009 5 week plan, 5 meals per day, 5 minute preparation time per meal, recipes with only 5 ingredients, 5 cheat days in 5 weeks, and 25 minute workouts 5 days a week for 5 weeks 1300 58 32 10 Weight Loss
The Abs DietDavid Zinczenko, Editor-in-Chief of Men’s HealthRodale Books, 2005

This diet is based on foundation foods that conform to the acronym Abs Diet Power :

  • A lmonds and other nuts
  • B eans and legumes
  • S pinach and green vegetables
  • D airy (fat free or low fat)
  • I nstant Oatmeal
  • E ggs
  • T urkey and lean meats
  • P eanut butter (natural and sugar free)
  • O live oil
  • W hole-grain breads and cereals
  • E xtra protein (whey powder)
  • R aspberries and other berries
1700 45 25 30 Weight Maintenance
Atkins™ for LifeDr. Robert C. Atkins™, MDSt. Martin's Griffin, 2004 A low carbohydrate plan for those who have lost weight with the original Atkins™ Diet. Dieters are advised to cut back on carbohydrates if weight loss stops. Phase 1: 1540 24 21 55 Weight Maintenance
Phase 2: 1970 22 22 56 Weight Maintenance
Phase 3: 2310Pre-Maintenance 29 19 52 Weight Maintenance
Phase 4: 2050Lifetime Maintenance 35 20 44 Weight Maintenance
Eat Right 4 Your Type (The Blood Type Diet)Dr. Peter J. D’AdamoPutnam Adult, 1996 Based on the idea that tailoring one’s diet based on blood type (A, B, O, AB) will result in weight loss and overall health. Blood Type O: 1000 44 29 27 Weight maintenanceOverall Health
Blood Type A: 1150 55 10 34
Blood Type B: 1200 55 22 23
Blood Type AB: 1200 56 25 20
Body for LifeBill Phillips, Michael D’OrsoWilliam Morrow, 1999 This book focuses primarily on exercise, and recommends 6 small meals per day for 6 weeks, consisting of lean meats, vegetables, whole grains, healthy fats, and fish in addition to strenuous exercise. 1270 45 45 10 Exercise and nutrition for quick weight loss
The New Cabbage Soup DietMargaret DanbrotSt. Martin's Paperbacks, 2004 Very-low calorie diet plan, based on the theory that monotony will cause the person to stop eating. Only food consumed is cabbage soup supplemented occasionally by specific fruits and vegetables 700 57 18 25 Weight Loss
The Cheaters DietPaul Rivas, MDHCI, 2005 Based on the plate method: 1/2 plate vegetables, 1/4 whole grains, 1/4 lean protein. Dr. Rivas claims that you must cheat on the weekends to “stroke your metabolism and boost fat loss.” He suggests eating “whatever you want” from 9am on Saturday to 9pm on Sunday. 1200-excessive calories 50 20 30 Weight Maintenance
The Diet Solutionhttp://www.thedietsolutionprogram.com/N/A Diet that promotes organic and “natural” foods, free of processing, regardless of the macronutrient composition. Excludes soy products. 928 34 20 46 Weight loss
Eat This, Not That!David Zinczenko with Matt GouldingRodale Books, 2009 Written by the editor-in-chief of Men’s Health magazine, aimed at male readers who eat mainly at fast food restaurants. Attempts to provide readers with alternative options to calorie laden fast food choices. - - - - Weight maintenance
Flat Belly DietLiz Vaccariello and Cynthia SassRodale Books, 2009 Premise is to trim calories to 1600, add a mono-unsaturated fatty acid at every meal, eat every four hours, and to perform regular exercise to lose weight and belly fat. 1600 42 25 33 Weight loss
French Women Don't Get FatMireille GuilianoVintage, 2007 Lifestyle changes illustrated through an autobiography of the author 1200-1300 43 22 45 Low Calorie
Change Your Genetic Destiny (The GenoType Diet)Dr. Peter J. D’Adamo with Catherine WhitneyBroadway, 2009 An expansion on the concept of the Blood Type Diet, created by naturopathic physician Dr. D’Adamo. Dr. D’Adamo identifies the six “GenoTypes”: the Hunter, the Gatherer, the Teacher, the Explorer, the Warrior, and the Nomad. Diet helps consumers map out their genetic makeup and discover which “GenoType” they are.The theory is that readers can reprogram their gene responses to lose and maintain weight, among other health improvements, by choosing foods that enhance each GenoType, and avoiding foods that do not. - - - - Weight maintenanceOverall health
The New Glucose Revolution: The Authoritative Guide to the Glycemic IndexJennie Brand-Miller, Phd; Thomas Wolever, MD, Phd; Kaye Foster-Powell; Stephen Colagiuri, MDMarlowe and Co., 2006 The theory is that simple carbohydrates cause spikes in blood sugar levels, causing recurrent hunger. Recommends eating low glycemic-index foods (i.e., whole grains, protein) to stave off hunger and weight gain. 1200 55 24 21 Weight Maintenance
LearnKelly BrownellAmerican Health Publishing Company, 2004 Lifestyle, exercise, attitudes, relationships and nutrition following government recommendations. 1650 55 15 30 Weight Maintenance
Mediterranean Recommends grains, vegetables, and sources of healthy fats (e.g., olive oil and nuts). - 45 20 35 Weight Maintenance
The Perricone PrescriptionDr. Nicholas PerriconeHarper Paperbacks, 2004 Anti-inflammatory foods eaten to reverse aging. Unlimited salmon. 1300 35 39 26 Anti-inflammatory/Overall Health
Pritikin Program for Diet and ExerciseNathan PritikinBantam, 1984 Six meals per day, no portion control. - 80 10 <10 Low fatVegetarianWeight loss
The Complete Scarsdale Medical DietDr. Herman TarnowerBantam, 1982 Artificial sweeteners and appetite suppressants are recommended. 1000 21 46 35 Weight loss
Sensa Weight-Loss Program (The Sprinkle Diet)Dr. Alan Hirsch, MD, FACPHilton Publishing, 2009 Flavorless sprinkles (“Tastanants”) sprinkled on food, helps dieters eat less and feel full faster. - - - - Weight loss
The Serotonin Power DietJudith J. Wurtman, PhDNina T. Frusztajer, MDRodale Books, 2006 The authors claim carbohydrate-rich snack eating will decrease stress and help dieters lose weight by producing more serotonin. 1500 62% 18% 20% Weight loss
The Sonoma DietConnie Guttersen RD, PhDMeredith Books, 2005 Influenced by a Mediterranean plant-based diet. This three phase diet places emphasis on a variety of flavorful, nutrient dense "power foods" such as almonds, bell peppers, blueberries, broccoli, grapes, olive oil, spinach, strawberries, tomatoes, and whole grains. 1500 for men 50-55 15-20 30 Low Calorie
1200 for women 50-55 15-20 30 Low Calorie
South Beach DietDr. Arthur AgatstonRodale, Inc., 2003 3 phase book by Arthur Agaston MD; Low carbohydrate*Phases based on a 140-pound, 40-year-old, lightly active woman Phase 1: 1850 16 38 46 Low Calorie
Phase 2: 1450 37 26 40 Low Calorie
Phase 3: 1750 31 29 40 Low Calorie
The Spectrum DietDr. Dean Ornish, MDBallantine Books, 2008 A lifestyle change; find where you fall on Ornish’s food “spectrum” (Group 1 being the healthiest, Group 5, the least healthy) and make changes according to your desired health outcomes (e.g., weight loss, weight maintenance, reduced risk of cancer, etc.).Plan calls for regular exercise (aerobic, resistance training, and flexibility), stress management (yoga, meditation), nutrition advice (low-fat, vegetarian), and nurturing relationships. 1580 70 20 10 Low-fat/vegetarian for overall improved health
Sugar BustersH. Leighton Steward; Morrison C. Bethea, MD; Sam S. Andrews, MD; Luis A. Balart, MDBallantine Books, 1999 Cut or completely eliminate dietary sugar to trim fat 1200 40 30 30 Weight loss
The Supermarket DietJanis Jibrin MS, RDHearst, 2007 Provides shopping lists, meal plans, recipes and snacks. The book begins with at two week boot camp phase. The author helps readers select which calorie level is the best fit for them, and how to troubleshoot problems if the calorie level does not seem to be yielding results. 1200-1500 50 20 30 Low CalorieWeight Maintenance
The Ultimate Weight SolutionDr. Phil McGrawFree Press, 2004 Dr. Phil McGraw authored this 3 phase diet book Phase 1: 1300 47 36 17 Low Calorie
Phase 2: 1100 49 32 19 Very low calorie
Phase 3: 1820 52 27 17 Weight Maintenance
The Volumetrics Weight-Control PlanBarbara Rolls, PhDHarperTorch, 2002 Focuses on satiety and feeling full, by filling up on high volume foods with low energy density (e.g., soup) 1700 61 23 18 Weight lossWeight maintenance
YOU: On a Diet (Revised Edition)Dr. Mehmet Oz and Dr. Michael RoizenFree Press, 2009 Weight loss with an emphasis on waist measurement and its relationship to health. 1700 46 21 33 Weight lossWight maintenance
The Zone DietDr. Barry SearsThorsons, 1999 Balances carbohydrates, protein, and fat to stabilize the hormones that trigger hunger and weight gain. 1700 40 30 30 Weight loss

10.5 Web-Based Treatment Programs and Resources

The Internet provides some excellent resources for those who want and need more information. However, it also includes sites with questionable recommendations, so individuals should proceed with caution. Web-based resources are discussed below.

10.6.1 Available Programs

Two types of programs are available. First are those that primarily provide information. Second are those that counsel the individual and provide low calorie diets and other advice. Sound Internet resources that can help those who are trying to control their weights are listed in Table 25. These fall into the category of informational resources. The reader needs to be aware that not all sites providing advice and information are sound. It is best to trust the sites sponsored by government, professional, and voluntary associations with some standing and expertise in the weight control field. A new entry into the weight loss arena in recent years is the web-based weight control program (210). These are examples of the second type of program. Resources include chat rooms, diet and exercise information, and often products that are for sale. For example, Nutrisystem®.com requires the purchase of prepackaged foods in order to access their web site. The ediets.com website, another weight control program, charges a monthly fee to use its site. It provides shopping lists from which consumers self-select foods, and also it provides general advice.

These web-based commercial offerings vary in their quality, some are very good and others are quite poor.

 

Table 25. Internet Resources for Weight Control (164-167)
Type of Site and Name Internet address and Comments
Advice and Information on nutrition and weight control
American Dietetic Association www.eatright.orgThis is the website for nutrition professionals. Membership allows entrance to the Journal of the American Dietetic Association and the Evidenced Based Library.
Shape Up! America www.shapeup.orgProvides good yet minimal nutrition education information for patients. Helpful sample menus are provided for 1500 and 2000 calorie diets as well as practical ways to increase physical activity.Founded in 1994, Shape Up America! is a 501(c)3 not-for-profit organization committed to raising awareness of obesity as a health issue and to providing responsible information on healthy weight management.
American Obesity Association www.obesity.orgComprehensive website dedicated to obesity. Provides helpful information for patients as well as links to other nutrition/fitness resources. This website also offers information on treatment, prevention, education, various aspects of public policy, and obesity research for professionals.
National Institutes of Health www.nutrition.govExtremely helpful resource for patients and health professionals alike. The website is home to the Dietary Guidelines, Mypyramid which is an interactive diet and physical activity planner, and an abundance of information regarding health and nutrition.
Weight-Control Information Network (WIN) http://win.niddk.nih.gov/A helpful information service of the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH) for health professionals and the public. WIN was established in 1994 to provide the general public, health professionals, the media, and Congress with up-to-date, science-based information on obesity, weight control, physical activity, and related nutritional issues.
Web-Based Help for Dieters
Cyberdiet.com www.cyberdiet.comThe program is $38.87 per 3 months or $77 annually via www.dietwatch.com. Resources on fitness, nutrition, motivation, wellness, recipes, and emotional health are included in each counseling plan. RDs provide the nutrition recommendations and accredited health professionals provide other health counseling/information.
WebMD Nutrition Resources http://www.webmd.com/diet/default.htmThis is an extremely useful resource for patients. The site provides a BMI calculator, calorie counter, diet evaluator, fitness and diet journal, a large food/nutrient database, a fiber calculator, helpful articles, videos, and slideshows on eating healthfully, and charts that can be personalized.
Ediets.com www.ediets.comProvides a Pyramid based reducing diet and food list, fitness information, healthy recipes, social networking community, and charts and dieting tools. Cost is currently $17.95 for the first 4 weeks. Meal delivery options are also available for $19.95 per day. This option includes 3 meals per day and a snack or dessert in addition to a social networking community and nutrition and fitness guidance.
Weight Watchers® Online www.weightwatchers.comSelf-help version of Weight Watchers® program provides a diet plan and fitness information with exercises. Cost is currently $53.85 per month for 3 months and $17.95 for each month after. Very consumer friendly.
Lifepractice.com www.lifepractice.comThe Life Practice program deals with exercise, nutrition, sleep habits and stress management. A personal coach is assigned to each member. Frequent e-mails and daily tracking of the member's progress is standard. The cost is $3.50 per week.
Fitday.com http://fitday.com/This website is a free online journal that tracks and analyzes food intake, exercise, and weight loss goals.
caloriescount.com www.caloriescount.comThis site boasts several online calculators and tools that help the participant keep track of their weight loss, such as the exercise/calories burned calculator and online diet meal plans given the participants appropriate caloric range, which may also be determined from the site. These tools are free of charge. Access.
SparkPeople® www.sparkpeople.comA comprehensive website with free nutrition, health, and fitness tools, support, and resources. This website boasts a large online social support network and may particularly help those with a preference towards online community networks.
Diet.com www.diet.comA nutrition and health website with tools to aid in weight loss, healthy living, and wellness. Basic membership is free and includes access to online support groups, articles, and useful tracking tools. However, premium membership, including all of the above in conjunction with a customized diet plan, personalized exercise plan and coping plan, personalized expert advice, and individualized weekly self checklists to help keep you on track, requires a $19.95 initiation fee and depending on the plan may range from $9.95 to $19.95 per month.

10.6.2 Candidates

Those who are overweight or moderately obese with few risk factors, and who need additional support and information after they have been screened by a physician on weight reduction, may find these resources useful. They are not freestanding and need to be administered in conjunction with some additional health and dietary counseling about a hypocaloric diet from a physician or registered dietitian.

10.6.3 Advantages

The Internet is widely available at all times of the day or night, at low cost. For example, the US Army has developed a web-based dietary advice program that can be used at Army bases around the world.

10.6.4 Disadvantages

High-risk patients, especially those who lack economic resources, may attempt to use these Internet sites for the primary treatment of their condition. Also, some sites provide inappropriate or wrong advice and there is little personal supervision or support of the dieter. Peer support networks and chat rooms can be beneficial to a patient trying to find support for weight loss, but there is also a risk that peers may provide incorrect advise or promote unsubstantiated weight loss techniques.

10.6.5 Safety and Effectiveness

The safety and effectiveness of internet sites for weight reduction has not been established (105). Only recommended sites should be trusted.

10.7 Voluntary Self-Help Programs

Self-help programs led by laypersons are voluntary programs that charge very low or no fees. National organizations include TOPS Club, Inc.® (Take Off Pounds Sensibly), OA (Overeaters Anonymous®), and others. These programs are designed primarily to provide group support to those who have weight problems, rather than to provide and supervise weight reduction diets.

10.8 Mobile Applications for Weight Loss

As people’s lives become more mobile, so do the tools that help them lose weight. Calorie tracking programs are available to monitor daily intake, and fitness programs provide sample workouts and activity logs. Other apps are available to promote water consumption and provide tips and support. As with web-based resources, these should be used in conjunction with physician or dietician-led interventions, and not as a replacement for them. Though research on the efficacy of these programs is still scant, one recent study suggests that combining some form of counseling alongside a smart-phone based platform led to greater results than either counseling or smart-phone use alone (237). Examples of current available apps and the platforms on which they are available are summarized in Table 26.

Table 26. Mobile Apps to Aid in Weight Loss
App Name Cost Available Platforms Features
My Fitness Pal free Apple, Android,
Windows, Blackberry, and online (http://www.myfitnesspal.com)
Features include a calorie tracker, activity tracker, weight tracker, body measurement tracker, water tracker, extensive food list including restaurants, barcode scanner, breakdown of intake including protein, fat, carbohydrates, fiber, and vitamins and minerals, daily and weekly summaries, ability to add friends, and compatibility with fitbit, map my fitness, and other apps and devices.
Lose It! Free (premium upgrade for $39.99/yr) Apple, Android, and online (https://www.loseit.com) Features include calorie tracker, activity
tracker, weight tracker, barcode
scanner, extensive food list with restaurant options, ability to add recipes, breakdown of intake by protein, fat, and carbohydrates, daily and weekly summaries by email, ability to add friends, compatibility with Nike+ fuelband. Premium features include additional compatibility with apps and devices, hydration and sleep trackers, body measurement tracker, and blood glucose
and blood pressure trackers.
Mapmyfitness free (premium upgrade for $29.99/yr for apple and online) Apple, Android, Blackberry (limited), and online (http://www.mapmyfitness.com) Features include allowing users to track a variety of activities in real time and after the fact, search for and save routes for walking, jogging, biking, and hiking, a food tracker, the ability to play music from music library on phone, add friends and share workouts. “Mapmyrun” and “Mapmyride” are related apps for running and biking respectively. Premium features include training plans,
training features, and live location tracking.
Hy free Apple Features include allowing users to set water goals in ml or fl oz, add water consumed in increments of 50 or 100 ml (2 or 4 fl oz), the ability to set reminders to consume water, and facts about the importance of water consumption displayed daily.
EaTipster free Apple Features include daily tips on healthy eating from dietitians, ability to save and share tips, and the ability to set reminder for tips

11. Summary of Weight Loss Phase

Current guidelines for the composition of weight reducing diets, as discussed above, are outlined in the 2010 Dietary Guidelines for Americans (Table 1), as well as the MyPlate.gov website.

12. Weight Maintenance Phase of Weight Control

Once obese individuals have lost weight, their healthier weight and current fat mass must be maintained. This is the weight maintenance phase of weight control. It involves alterations in dietary intake and physical activity from levels that are different to those at the onset of dietary treatment. Energy needs are lower to stay in energy balance than they were prior to weight reduction, even though weight was lost. This is because both fat and lean body mass is lost during the weight reduction phase. With a loss of lean body mass comes a decrease in metabolically active tissue, which then reduces resting metabolism. In addition, it takes less effort for an individual to move with a now lighter body, so the energy cost of physical activity is reduced. The implications are that a slight decrease in energy intake from prior levels and an increase in energy output is necessary during weight maintenance. There is a need for continued attention to these factors by both the physician and patient. Behavior modification is necessary to sustain lifestyle changes developed during the weight reduction phase. It is best initiated during the weight loss phase, and maintained thereafter. All too often the weight maintenance phase is neglected or ignored, and weight is regained over the long term (71). Some factors that seem to be associated with long-term successful weight maintenance include continued regular exercise and to a lesser extent, use of low calorie, low fat diets relatively high in fruits and vegetables. Also, continued self-monitoring of the amount and type of food consumed and of physical activity levels may help (71).

12.1 Nutrient Needs

Although energy needs are less during weight maintenance, the requirements for protein, essential fatty acids, carbohydrate, dietary fiber, vitamins and minerals are similar to those of any normal adult. The 2010 Dietary Guidelines for Americans recommend that all Americans, including those who are watching their weight, to adopt the habits listed (see Table 1).

There is currently much debate about the ideal macronutrient distribution in diets for weight maintenance, but at present very few long-term studies are available on the effects of macronutrient content on weight maintenance. A low-fat, reduced-energy is the best studied diet and the most prescribed for weight maintenance (AND).

 

12.1.2 Carbohydrate

A recent study of the self-selected diets of free-living American adults found that diets high in carbohydrates (above 55% of calories) were lower in total energy and were associated with lower BMI’s than those consuming fewer carbohydrates. The nutrient density (amount of the nutrient per calorie consumed) of those with higher carbohydrate intakes was also higher for vitamins A, Vitamin C, carotene, folate, calcium, magnesium, and iron, but lower in vitamin B-12 and zinc than those with a lower intake of carbohydrates. Also, the high carbohydrate group ate more low-fat foods, grain products and fruits in addition to lower sodium intakes (214). Although individuals who choose to go on a very-low-carbohydrate diet may see increased weight loss within the first six months, these results are not sustained at 12 months (241). It is thought that this diet is not sustainable for long-term weight maintenance and instead, the diet should be a slight reduction in energy all together as well as an increased focus on the reduction of fat (241).

12.1.3 Energy Density

Other studies suggest that energy density of the diet rather that the macronutrient composition of the diet affects energy intake the most (215;216). One review found that low fat, high fiber diets were the most effective in promising weight loss, and that their effects appeared to be associated with energy density (217). Whether this is true in weight maintenance remains to be determined. There may be macronutrient effects on hunger and satiety that operate through endocrine and metabolic mechanisms such as leptin, insulin, grhelin, adiponectin and other hormones, which are only now being discovered. These hormones regulate food intake and may be altered. Additionally, the macronutrient composition of habitual diets also affects health risks. Finally, psychological and behavioral factors may make different macronutrient combinations more acceptable to some people. Currently, these topics are the subject of much debate, but research is needed to clarify what and which nutrient composition is optimal. What is currently recommended by the Academy of Nutrition and Dietetics with a strong rating is that an individualized reduced calorie diet is imperative for weight maintenance. By reducing fat, an individual is able to cut out more calories but it is suggested that both fat and carbohydrate be decreased (241).

Other forms of low energy dense diets would be the use of meal replacements or very-low-energy diets. Using meal replacements can be helpful for those who have trouble planning and preparing meals. They can also be helpful for those who experience a large amount of anxiety during meal times. An individual is able to replace one or two meals or snacks with these meal replacements with known nutrient content to help them stay at a calorie deficit of 500 – 1000 cal/day (241). This is something that could be used periodically to help patients get back on track after a relapse of poor food choices and is something that can be maintained long-term. Very-low-energy diets use meal replacement bars or shakes as the sole source of energy during the weight loss phase, however, this is not suitable for weight maintenance and instead used for quick weight loss and requires additional medical monitoring (241).

12.1.3 Fat

Although much remains to be discovered about the optimal dietary pattern for weight maintenance, a strong case can be made for keeping dietary fat levels below 30% of calories. In studies in which dietary fat was reduced from 35 to 25% of calories with no other recommendations, energy intake was reduced and weight was lost (218). It was estimated that reducing fat by 10% to within the range of 20 to 30% of calories would result in a loss of about 16 grams of body fat a day as a result of reduced energy intake. However, moderation in caloric intake is also necessary. Studies of free-living humans ranging from dietary changes produced only modest body weight losses of about one to three kilograms (182;219;220). Low fat diets consumed on an ad-libitum basis tend to be high in carbohydrate, but LDL cholesterol decreases, plasma triglycerides tend to normalize, and so do HDL/total cholesterol ratios (221). Finally, weight control may be easier (222).

12.1.4 Dietary Fiber

Although the influence of dietary fiber on energy regulation is still not clear, there is evidence that increased dietary fiber intake of about 15 grams appears to be associated with decreased energy intake and body weight of about two kilograms over several months. These effects may even be greater in overweight persons (181). However, these effects are not yet confirmed. Since dietary fiber intake is currently low, only about 15 grams per day in most Americans, and recommendations are for nearly twice that much, increased fiber levels seem to be appropriate, regardless.

12.1.5 Vitamins and Minerals

It is important to meet the current dietary recommendations of 3 – 4 servings of low-fat or non-fat dairy in order to get the daily suggested values of calcium and vitamin D. Research suggests that those with lower calcium intake have increased body weight (241). However, the mechanism by which this works is still unclear.

12.1.6 Lessons from Long Term Maintainers: Importance of Increased Physical Activity

Long-term follow-up of health outcomes demonstrates the need for permanent changes in weight toward healthier levels. The relative lack of effects of temporary downward fluctuations underscores the need for long-term weight maintenance. Data collected from individuals successful at weight loss and maintenance have enhanced our understanding of the most effective strategies in the long-term maintenance of healthier weights and prevention of relapse. Attention to moderation in dietary intake and the maintenance of high levels of physical activity is vital (223-226). Behavioral and attitude adjustments are also important. Encouraging data suggest that behaviors associated with maintenance of weight loss require less effort and become more pleasurable over time (29). In a recent telephone survey, 48% of individuals who had ever lost more than 10% of their body weight had maintained this loss for at least one year, and 26% had maintained for at least 5 years (227). Although these data are self-reported, they suggest progress in the avoidance of relapse and weight gain.

13. Adopt a Long-Term Eating Pattern to Maintain Weight: Adequate Nutrients within Calorie Needs

Most individuals appear to be aware of and use recommended measures, such as increased physical activity, decreased fat intake, decreased food portions, and decreased energy intakes. The problem is that they do so, but not for enough of the time. However, it is also true that dieting efforts often fail, and weight is often rapidly regained, probably negating predicted health benefits. Chronic dieters tend to be food-preoccupied, distractible, emotional, binge-prone, and unhappy, particularly when the diets are very restrictive (228). It is thus important to foster a healthy, balanced, stable relationship with food and diet. Health professionals can play a vital role in helping patients develop such a relationship.

14. Conclusions: Is Dieting Worth It?

About 39% of women and 21% of men in Western countries have ever tried to lose weight, and approximately 24% of women and 8% of men report that they currently are on a “diet”. In contrast, about 25% of men and 30% of women report that they are watching what they eat to avoid weight gain or to maintain their weights at current levels (229). Hypocaloric diets to induce loss of body fat therefore appear to be a common component of the weight control efforts of many people.
These realities and disadvantages have rightly led to questions about whether dieting is “worth it”, and whether the treatment is worse than the disease. Certainly they suggest that quality of life measures should be included in studies of reducing diets.
This chapter has stressed the role of the dietary treatment of obesity as a part of a comprehensive program of weight control that includes increased physical activity, lifestyle modification, appropriate intakes of nutrients to minimize chronic disease risk, and eating patterns that maximize quality of life. Such dietary treatment in those with mild to moderate obesity helps to decrease risk factors relative to baseline weights after five years. Therefore some health benefit, although it is limited, may be present. However, the health risk/benefit may be negative when dieting entails a cycle of rapid loss followed by equally rapid weight gain. From the standpoint of quality of life and mental health, psychosocial problems do not appear to be inevitable accompaniments of weight loss (184). Therefore, on balance, dietary approaches to obesity management do appear to be worthwhile, if and only if they are viewed as only one component of a long-term weight control program to keep weights and risks at healthier levels. Weight control is “Worth it”!

 

References

1. Fryar CD, Carroll mD, Ogden CL, Prevalence of Overweiht, Obesity and Extremem Obesity Among Adults: United States, Trends 1960-1962 Though 2009-2010. Division of Health and Nutrition Examination Surverys, 2012.
2. Oster G, Thompson D, Edelsberg J, Bird A, Colditz A. Lifetime Health and Economic Benefits of Weight Loss Among Obese Persons, American Journal of Public Health 1999, 89:10 (1536 – 1542).
3. Clinical Guidelines on the Identification, Evaluation, and Treatment of Overweight and Obesity in Adults--The Evidence Report. National Institutes of Health. Obes Res 1998; 6 Suppl 2:51S-209S.
4. Hill JO. Dealing with obesity as a chronic disease. Obes Res 1998; 6 Suppl 1:34S-38S.
5. Cummings S, Parham ES, Strain GW. Position of the American Dietetic Association: weight management. J Am Diet Assoc 2002; 102(8):1145-1155.
6. Kushner RF, Kus8.4hner N, Jackson Blatner D. Counseling Overweight Adults: The Lifestyle Patterns Approach and Toolkit. Chicago. American Dietetic Association, 2009.
7. Are Healthcare Professionals Advising Obese Patients to Lose Weight? A Trend Analysis, Medscape General Medicine CME
8. Goldstein DJ. Beneficial health effects of modest weight loss. Int J Obes Relat Metab Disord 1992; 16(6):397-415.
9. Estruch R, Martinez-Gonzalez MA, Corella D et al. Effects of a Mediterranean-style diet on cardiovascular risk factors: a randomized trial. Ann Intern Med 2006; 145(1):1-11.
10. Melanson KJ. CE Test: Food Intake Regulation in Body Weight Management: A Primer. Nutrition Today 2004; 39(5).
11. Natural Marketing Institute. 2006. Morleyville Pennsylvania; in published report.
12. Galuska DA, Will JC, Serdula MK, Ford ES. Are health care professionals advising obese patients to lose weight? JAMA 1999; 282(16):1576-1578.
13. The Obesity Epidemic: A Mandate for a Multidisciplinary Approach. Proceedings of a roundtable. Boston, Massachusetts, USA. October 27, 1997. J Am Diet Assoc 1998; 98(10 Suppl 2):S1-61.
14. Simkin-Silverman LR, Wing RR. Management of obesity in primary care. Obes Res 1997; 5(6):603-612.
15. U.S.Department of Health and Human Services and U.S.Department of Agriculture. Dietary Guidelines for Americans. 7th Edition. 2010. Washington, DC, U.S. Government Printing Office. 2010.
16. Committee to Reexamine IOM Pregnancy Weight Guidelines. Weight Gain During Pregnancy: Reexamining the Guidelines. Washington, D.C.: The National Academies Press, 2009.
17. Sacks FM, Svetkey LP, Vollmer WM et al. Effects on blood pressure of reduced dietary sodium and the Dietary Approaches to Stop Hypertension (DASH) diet. DASH-Sodium Collaborative Research Group. N Engl J Med 2001; 344(1):3-10.
18. U.S.Department of Health and Human Services, U.S.Department of Agriculture. Physical Activity Guidelines for Americans. 2008. Washington, DC, U.S. Government Printing Office.
19. Weight-control Information Network. Overweight and Obesity Statistics. National Institute of Diabetes and Digestive and Kidney Diseases. 1-6. 2012.
20. Fryar CD, Carroll MD, Ogden CL. Prevalence of overweight, obesity, and extreme obesity among adults: United States, trends 1960-1962 through 2009-2010. National Center for Health Statistics. 1-8. 2012.

21. Odgen CL, Carroll MD, Kit BK, Flegal KM. Prevalence of childhood and adult obesity in the United States, 2011-2012. JAMA 2014; 311(8):806-814.

22. Finkelstein EA, Trogdon JG, Cohen JW, Dietz W. Annual medical spending attributable to obesity: payer-and service service-specific estimates. Health Aff 2009. 28(5):w822-w831

23. 2013 AHA/ACC/TOS guidelines for the management of overweight and obesity in adults: A report for the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and The Obesity Society. JACC 2014. 63(25):2985-3023.

24. Clinical Guidelines on the Identification, Evaluation, and Treatment of Overweight and Obesity in Adults--The Evidence Report. National Institutes of Health. Obes Res 1998; 6 Suppl 2:51S-209S.
25. Weight-control Information Network. Statistics Related to Overweight and Obesity. National Institute of Diabetes and Digestive and Kidney Diseases, editor. 1-8. 2007.
26. Miles JM, Jensen MD. Counterpoint: visceral adiposity is not causally related to insulin resistance. Diabetes Care 2005; 28(9):2326-2328.
27. Lebovitz HE, Banerji MA. Point: visceral adiposity is causally related to insulin resistance. Diabetes Care 2005; 28(9):2322-2325.
28. Kushner RF, Blatner DJ. Risk assessment of the overweight and obese patient. J Am Diet Assoc 2005; 105(5 Suppl 1):S53-S62.
29. Klem ML, Wing RR, Lang W, McGuire MT, Hill JO. Does weight loss maintenance become easier over time? Obes Res 2000; 8(6):438-444.
30. Treatment of the Obese Patient. Totowa: Humana Press, 2007.
31. Jensen MD. Expert panel report: Guidelines (2013) for the management of overweight and obesity in adults. Obesity 2014; 22:S41-S410. Doi:10.1002/oby.20660.
32. Klein S, Allison DB, Heymsfield SB et al. Waist circumference and cardiometabolic risk: a consensus statement from shaping America's health: Association for Weight Management and Obesity Prevention; NAASO, the Obesity Society; the American Society for Nutrition; and the American Diabetes Association. Diabetes Care 2007; 30(6):1647-1652.
33. Haven J, Britten P. MyPyramid-The Complete Guide. Nutrition Today 2006; 41(6).
34. Latner JD. Self-help for Obesity and Binge Eating. Nutrition Today 2007; 42(2).
35. Weight-control Information Network, National Institute of Diabetes and Digestive and Kidney Diseases, U.S.Department of Health and Human Services, National Institutes of Health. Prescription Medications for the Treatment of Obesity. 2013.

36. U.S. Food and Drug Administration. FDA approves weight-management drug Contrave. U.S. Department of Health and Human Services. 11 September, 2014. Available at: http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm413896.htm.

37. Weight-control Information Network, National Institute of Diabetes and Digestive and Kidney Diseases, U.S.Department of Health and Human Services, National Institutes of Health. Prescription Medications for the Treatment of Obesity. 2007.
38. Fujioka K, Lee MW. Pharmacologic treatment options for obesity: current and potential medications. Nutr Clin Pract 2007; 22(1):50-54.
39. American Dietetic Association. Managing Obesity: A Clinical Guide. Second ed. Diana Faulhaber, American Dietetic Association, 2009.
40. I. Moyers SB. Medications as adjunct therapy for weight loss: approved and off-label agents in use. J Am Diet Assoc 2005;105(6):949-959.
41. Palamara KL, Mogul HR, Peterson SJ, Frishman WH. Obesity: New Perspectives and Pharmacotherapies. Cardiology in Review 2006; 14(5).
42. Genentech USA I. About Xenical: Mechanism of Action. 2009.
43. European Medicines Agency (EMEA). Public Statement on Acomplia (Rimonabant): Withdrawal of the Marketing Authorisation in the European Union. Public Statement. In press.
44. U.S. Food and Drug Administration. Meridia (sibutramine): Market Withdrawal Due to Risk of Serious Cardiovascular Events. U.S. Department of Health and Human Services. 08 October, 2014. Available at: http://www.fda.gov/safety/medwatch/safetyinformation/safetyalertsforhumanmedicalproducts/ucm228830.htm.

45. U.S. Food and Drug Administration. Tainted weight loss products. U.S. Department of Health and Human Services. 12 September, 2014. Available at: http://www.fda.gov/Drugs/ResourcesForYou/Consumers/BuyingUsingMedicineSafely/MedicationHealthFraud/ucm234592.htm.
46. Kolasa KM, Kay C, Henes S, Sullivan C. The clinical nutritional implications of obesity and overweight. N C Med J 2006; 67(4):283-287.
47. L. Weight and Wellness Center at Tufts Medical Center. Guide for Eating After Gastric Bypass Surgery. Available at: https://www.tuftsmedicalcenter.org/~/media/Brochures/TuftsMC/Patient%20Care%20Services/Departments%20and%20Services/Weight%20and%20Wellness%20Center/GBP%20Diet%20Manual12611.ashx
48. Medscape General Medicine CME. 2005; 7(4): 10
49. Carvajal R, Wadden TA, Tsai AG, Peck K, Moran CH. Ann. N.Y. Acad. Sci. 2013 191-206. doi: 10.111/nyas. 12004
50. Thande NK, Hurstak EE, Sciacca RE, Giardina EGV. Obesity, 2009; 17(1); 107-113. doi: 10.1038/oby.2008.478

51. Serdula MK, Mokdad AH, Williamson DF, Galuska DA, Mendlein JM, Heath GW. Prevalence of attempting weight loss and strategies for controlling weight. JAMA 1999; 282(14):1353-1358.
52. Battle EK, Brownell KD. Confronting a rising tide of eating disorders and obesity: treatment vs. prevention and policy. Addict Behav 1996; 21(6):755-765.
53. Melanson KJ, Angelopoulos TJ, Nguyen VT et al. Consumption of whole-grain cereals during weight loss: effects on dietary quality, dietary fiber, magnesium, vitamin B-6, and obesity. J Am Diet Assoc 2006; 106(9):1380-1388.
54. Van D, Lindley EM. Cognitive and behavioral approaches in the treatment of obesity. [Review] [131 refs]. Endocrinology & Metabolism Clinics of North America 2008; 37(4):905-922.
55. Weight Control Information Network. 2012
56. National Institues of Health. Talking with Patients about Weight Loss: Tips for Primary Care Professionals. NIH Publication No. 07-5634. 2008.
57. American Dietetic Association (ADA). Adult Weight Management Guideline: Major Recommendations. ADA Evidence Analysis Library . 2009.
58. Pinto AM, Gorin AA, Raynor HA, Tate DF, Fava JL, Wing RR. Obesity, 2008; 16(11): 2456-2461. doi: 10.1038/oby.2008.364
59. Koplan JP, Dietz WH. Caloric imbalance and public health policy. JAMA 1999; 282(16):1579-1581.
60. National Heart LaBI. Guidelines on Overweight and Obesity: Electronic Textbook. 1995.
61. Fabricatore AN, Wadden TA, Womble LG et al. The role of patients' expectations and goals in the behavioral and pharmacological treatment of obesity. Int J Obes (Lond) 2007; 31(11):1739-1745.
62. Foster GD, Wadden TA, Vogt RA, Brewer G. What is a reasonable weight loss? Patients' expectations and evaluations of obesity treatment outcomes. J Consult Clin Psychol 1997; 65(1):79-85.
63. Dwyer JT, Dionne D, Stevens A. Popular and Fad Diet Programs: Nutritional Adequacy, Safety, and Efficacy. In: Latner J, Wilson T, editors. Self-Help Approaches for Obesity and Eating Disorders. New York: The Guilford Press, 2007: 21-52.
64. U.S.Department of Health and Human Services, National Institues of Health, National Heart LaBI. Aim for a Healthy Weight. NIH Publication No. 05-5213. 2005.
65. National Institutes of Health Obesity Education Initiative. The Practical Guide Identification, Evaluation, and Treatment of Overweight and Obesity in Adults. 00-4084. 2000. National Institues of Health, U.S Department of Health and Human Services, Public Health Services, National Institutes of Health, National Heart, Lung, and Blood Institute.
66. Provencher V, Begin C, Tremblay A et al. Health-at-every-size and eating behaviors: 1-year follow-up results of a size acceptance intervention. J Am Diet Assoc 2009; 109(11):1854-1861.
67. Wildman REC, Miller BS. Sports and Fitness Nutrition. Australia: Thomson/Wadsworth, 2004.
68. Melanson KJ. CE Test: Food Intake Regulation in Body Weight Management: A Primer. Nutrition Today 2004; 39(5).
69. Melanson KJ. CE Test: Food Intake Regulation in Body Weight Management: A Primer. Nutrition Today 2004; 39(5).
70. Seagle HM, Strain GW, Makris A, Reeves RS. Position of the American Dietetic Association: weight management. J Am Diet Assoc 2009; 109(2):330-346.
71. Brownwell KD, Fairburn CG. Eating Disorders and Obesity: a Comprehensive Handbook. New York: Guilford Press, 2005.
72. Dwyer JT. Treatment of Obesity: Conventional Programs and Fad Diets. In: P.Bjorntorp, B.N.Bordoff, editors. Obesity. New York: Lippincott, 1992: 662-676.
73. Chaston TB, Dixon JB, O'Brien PE. Changes in fat-free mass during significant weight loss: a systematic review. Int J Obes (Lond) 2007; 31(5):743-750.
74. Nicklas JM, Huskey KW, Davis RB, Wee CC. Am J Prev Med. 2012; 42(5): 481-485. doi: 10.1016/j.amepre.2012.01.005
75. Fontanarosa PB. Patients, physicians, and weight control.[comment]. JAMA 1999; 282(16):1581-1582.
76. Hall KD, What is the Required Energy Deficit per Unit Weight Loss? Int J Obes (Lond). 2008; 32(3): 573-576
77. Rolls BJ, Drewnowski A, Ledikwe JH. Changing the energy density of the diet as a strategy for weight management. J Am Diet Assoc 2005; 105(5 Suppl 1):S98-103.
78. American Dietetic Association (ADA). Self-Monitoring. 2009. 11-23-2009.
79. Ness-Abramof R, Apovian CM. Diet modification for treatment and prevention of obesity. Endocrine 2006; 29(1):5-9.
80. Handbook of Assessment Methods for Eating Behaviors and Weight-Related Problems (Measures, Theory, and Research). Los Angeles, London, New Delhi, Singapore, Washington DC: Sage, 2009.
81. Burke LE, Swigart V, Warziski TM, Derro N, Ewing LJ. Experiences of self-monitoring: successes and struggles during treatment for weight loss. Qual Health Res 2009; 19(6):815-828.
82. Truby H, Baic S, deLooy A et al. Randomised controlled trial of four commercial weight loss programmes in the UK: initial findings from the BBC "diet trials". BMJ 2006; 332(7553):1309-1314.
83. Johnson RK, Soultanakis RP, Matthews DE. Literacy and body fatness are associated with underreporting of energy intake in US low-income women using the multiple-pass 24-hour recall: a doubly labeled water study. J Am Diet Assoc 1998; 98(10):1136-1140.
84. de JL, DeLany JP, Nguyen T et al. Validation study of energy expenditure and intake during calorie restriction using doubly labeled water and changes in body composition. Am J Clin Nutr 2007; 85(1):73-79.
85. Heshka S., Heymsfield S.B., Matthews DE. Doubly labeled water measures energy use. Science and Medicine 1994; 1(1):74.
86. Philippaerts RM, Westerterp KR, Lefevre J. Doubly labelled water validation of three physical activity questionnaires. Int J Sports Med 1999; 20(5):284-289.
87. Bonnefoy M, Normand S, Pachiaudi C, Lacour JR, Laville M, Kostka T. Simultaneous validation of ten physical activity questionnaires in older men: a doubly labeled water study. J Am Geriatr Soc 2001; 49(1):28-35.
88. HARRIS TJ, OWEN CG, VICTOR CR, ADAMS RIKA, EKELUND ULF, COOK DG. A Comparison of Questionnaire, Accelerometer, and Pedometer: Measures in Older People. [Miscellaneous Article]. Medicine & Science in Sports & Exercise 2009; 41(7):1392-1402.
89. Melanson EL, Knoll JR, Bell ML et al. Commercially available pedometers: considerations for accurate step counting. Preventive Medicine 2004; 39(2):361-368.
90. Bouten CV, Verboeket-van de Venne WP, Westerterp KR, Verduin M, Janssen JD. Daily physical activity assessment: comparison between movement registration and doubly labeled water. J Appl Physiol 1996; 81(2):1019-1026.
91. Konnikoff K, Dwyer J. Popular Diets and Other Treatments of Obesity. In: Lockwood D.H., Heffner T.G, editors. Popular Diets and Other Treatments of Obesity. Heidelberg and Berlin: Springer Verlag, 2000: 195-236.
92. Caputo FA, Mattes RD. Human dietary responses to covert manipulations of energy, fat, and carbohydrate in a midday meal. Am J Clin Nutr 1992; 56(1):36-43.
93. Saltzman E, Dallal GE, Roberts SB. Effect of high-fat and low-fat diets on voluntary energy intake and substrate oxidation: studies in identical twins consuming diets matched for energy density, fiber, and palatability. Am J Clin Nutr 1997; 66(6):1332-1339.
94. Blundell JE. The biology of appetite. Clinical Applied Nutrition 1991; 1:21-31.
95. Van Itallie T. Dietary approaches to the treatment of obesity. In: Stunkard A, editor. Obesity. Philadelphia: WB Sanders, 1980: 249-261.
96. Adam-Perrot A, Clifton P, Brouns F. Low-carbohydrate diets: nutritional and physiological aspects. Obes Rev 2006; 7(1):49-58.
97. Rosales-Velderrain, Armando; Goldberg, Ross F.; Ames, Gretchen E.; Stone, Ronald L.; Lynch, Scott A.; Bowers, Steven P. The American Surgeon 2014. 80(3): 290-294
98. St-Onge MP RFJAJHS. A new hand-held indirect calorimeter to measure postprandial energy expenditure. Obes Res 2004; 12(4):704-709.
99. Hall KD. Modeling metabolic adaptations and energy regulation in humans. Annu Rev Nutr. 2012 Aug 21;32:35-54. doi: 10.1146/annurev-nutr-071811-150705. Epub 2012 Apr 23.

100. Johannsen DL, Knuth ND, Huizenga R, Rood JC, Ravussin E, Hall KD. Metabolic slowing with massive weight loss despite preservation of fat-free mass. JJ Clin Endocrinol Metab. 2012 Jul;97(7):2489-96. doi: 10.1210/jc.2012-1444. Epub 2012 Apr 24.

101. all KD, Sacks G, Chandramohan D, Chow CC, Wang YC, Gortmaker SL, Swinburn BA. Quantification of the effect of energy imbalance on bodyweight. HLancet. 2011 Aug 27;378(9793):826-37. doi: 10.1016/S0140-6736(11)60812-X.

102. Freedman MR, King J, Kennedy E. Popular Diets: a Scientific Review. Obes Res 2001; 9(Suppl 1):1S-40S.
103. Tsai AG, Wadden TA. The evolution of very-low-calorie diets: an update and meta-analysis. Obesity (Silver Spring) 2006; 14(8):1283-1293.
104. National Task Froce on the Prevention and Treatment of Obesity. Very low-calorie diets. JAMA 1993; 270:967-974.
105. Tsai AG, Wadden TA. Systematic review: an evaluation of major commercial weight loss programs in the United States. Ann Intern Med 2005; 142(1):56-66.
106. Brehm BJ, Seeley RJ, Daniels SR, D'Alessio DA. A randomized trial comparing a very low carbohydrate diet and a calorie-restricted low fat diet on body weight and cardiovascular risk factors in healthy women. J Clin Endocrinol Metab 2003; 88(4):1617-1623.
107. Sours HE, Frattali VP, Brand CD et al. Sudden death associated with very low calorie weight reduction regimens. Am J Clin Nutr 1981; 34(4):453-461.
108. Wadden TA, B. RI. Very low calorie diets. In: Fairburn C, Brownell KD, editors. Eating disorders and obesity. New York and London: Guilford Press, 2002: 534-538.
109. National Institute of Diabetes and Digestive and Kidney Diseases. Very Low-calorie Diets. 2008. Weight-control Information Network.
110. Varady K, Bhutani S, Church EC, Klempel MC. Short-term modified alternate-day fasting: a novle dietary strategy for weight loss and cardioprotection in obese adults. American Journal of Clinical Nutrition 2009; 90(5):1138-1143.
111. Klempel MC, Bhutani S, Fitzgibbon M, Freels S, Varady KA. Dietary and physical activity adaptations to alternate day modified fasting: implications for optimal weight loss. Nutr J 2010; 9:35. Doi:10.1186/1475-2891-9-35.

112. Bhutani S, Klempel MC, Kroeger CM, Trepanowski JF, Varady KA. Alternate day fasting and endurance exercise combine to reduce body weight and favorably alter plasma lipids in obese humans. Obesity 2013; 21(7):1370-9. Doi:10.1002/oby.20353.

113. Eshghinia S, Mohammadzadeh F. The effects of modified alternate-day fasting diet on weight loss and CAD risk factors in overweight and obese women. J Diabetes Metab Disord 2013; 12(1):4. Doi:10.1186/2251-6581-12-4.

114. Klemsdal TO, Holme I, Nerland H, Pedersen TR, Tonstad S. Effects of a low glycemic load diet versus a low-fat diet in subjects with and without the metabolic syndrome. Nutr Metab Cardiovasc Dis 2010; 20(3):195-201. Doi:10.1016/j.numecd.2009.03.010.

115. Klempel MC, Kroeger CM, Varady KA. Alternate day fasting increases LDL particle size independently of dietary fat content in obese humans. Eur J Clin Nutr 2013; 67(7):783-5. Doi:10.1038/ejcn.2013.83.
116. Nordmann AJ, Nordmann A, Briel M et al. Effects of low-carbohydrate vs low-fat diets on weight loss and cardiovascular risk factors: a meta-analysis of randomized controlled trials. Arch Intern Med 2006; 166(3):285-293.
117. U.S. Department of Health and Human Services. Very low-calorie diets. Version current December 2012. Internet: http://www.win.niddk.nih.gov/publications/PDFs/verylowcaldietsbw.pdf (accessed 6/15/2014).
U.S. Dietary Guidelines for Americans: http://www.health.gov/dietaryguidelines/2015.asp

118. Johnston BC, Kanters S, Bandayrel K, Wu P, Naji F, Siemieniuk RD, Ball GDC, Busse JW, Thorlund K, Guyatt G, Jansen JP, Mills EJ. Comparison of Weight Loss Among Named Diet Programs in Overweight and Obese Adults: A Meta-analysis. JAMA. 2014;312(9):923-933. Doi:10.1001/jama.2014.10397.

119. Melanson KJ, Summers A, Nguyen V, Brosnahan J, Lowndes J, Angelopoulos TJ, Rippe JM. Body composition, dietary composition, and components of metabolic syndrome in overweight and obese adults after a 12-week trial on dietary treatments focused on portion control, energy density, or glycemic index. Nutr J 2012; 11:57. Doi:10.1186/1475-2891-11-57.

120.Sacks FM, Bray GA, Carey VJ et al. Comparison of weight-loss diets with different compositions of fat, protein, and carbohydrates. N Engl J Med 2009; 360(9):859-873.
121. Sacks F, Bray G, Carey V et al. Comparison of Weight-Loss Diets with Different Compositions of Fat, Protein, and Carohydrates. The New England Journal of Medicine 2009; 360(9):859-873.
122. Wadden TA, Bryne K, Krauthamer-Ewing S. Obesity: Management. In: Shils M, Shike M, Olsen J, Ross C, editors. Modern Nutrition in Health and Disease. Baltimore: Lippincott, Williams, and Wilkons, 2006: 1029-1042.
123. Foster GD, Makris AP. Low-carbohydrate Diets in the Treatment of Obesity. Nutrition Today 2005; 40(4).
124. Schoeller DA, Buchholz AC. Energetics of obesity and weight control: does diet composition matter? J Am Diet Assoc 2005; 105(5 Suppl 1):S24-S28.
125. World Health Organization. Evaluation of certain food additives and contaminants. 896, 1-128. 2000. Tech Rep Ser.
126. Dukan, P. The Dukan DIet. New York: Crown Archetype, 2011.
127. Atkins RC. Dr. Atkins' Diet Revolution. New York: Collins, 2002.
128. Eades M. Protein Power. New York: Bantum Books, 1996.
129. Gardner CD, Kiazand A, Alhassan S et al. Comparison of the Atkins, Zone, Ornish, and LEARN diets for change in weight and related risk factors among overweight premenopausal women: the A TO Z Weight Loss Study: a randomized trial. JAMA 2007; 297(9):969-977.
130. Steward H, Bethea MC, Andrews SS, Balart LA. Sugar Busters. New York: Ballantine Books, 1998.
131. Stubbs R, van Wyk M, Johnstone A, Harbron C. Breakfasts high in protein, fat or carbohydrate: effect on within-day appetite and energy balance. European Journal of Clinical Nutrition 1996; 50(7):409-417.
132. Crovetti R, Porrini M, Santangelo A, Testolin G. The influence of thermic effect of food on satiety. European Journal of Clinical Nutrition 1998; 52(7):482-488.
133. Eisenstein J, Roberts SB, Dallal G, Saltzman E. High-protein weight-loss diets: are they safe and do they work? A review of the experimental and epidemiologic data. Nutr Rev 2002; 60(7 Pt 1):189-200.
134. Nutrition recommendations and principles for people with diabetes mellitus. Diabetes Care 2000; 23 Suppl 1:S43-S46.
135. Grundy SM, Hansen B, Smith SC, Jr., Cleeman JI, Kahn RA. Clinical management of metabolic syndrome: report of the American Heart Association/National Heart, Lung, and Blood Institute/American Diabetes Association conference on scientific issues related to management. Circulation 2004; 109(4):551-556.
136. Klein S, Sheard NF, Pi-Sunyer X et al. Weight management through lifestyle modification for the prevention and management of type 2 diabetes: rationale and strategies: a statement of the American Diabetes Association, the North American Association for the Study of Obesity, and the American Society for Clinical Nutrition. Diabetes Care 2004; 27(8):2067-2073.
137. Pritikin M, McGrady PM. The Pritikin Program for Diet and Exercise. New York: Bantum Books, 1980.
138. Ornish D. Diet and fitness changes reverse coronary artery disease. World Review Nutrition and Dietetics 1993; 77:38-48.
139. Heller RF, Heller RF. The Carbohydrate Addict's Diet: the Lifelong Solution to Yo-Yo Dieting. New York: The Penguin Group, 1992.
140. Bernstein RK. Dr. Bernstein's Diabetes Solution: Complete Guide to Achieving Normal Blood Sugars. New York: Little, Brown, and Company, 2003.
141. Allan CB, Lutz W. Life Without Bread: How a Low-carbohydrate Diet Can Save Your Life. Los Angeles: Keats Publishing, 2000.
142. PENNINGTON AW. Treatment of obesity with calorically unrestricted diets. J Clin Nutr 1953; 1(5):343-348.
143. Surwit RS, Feinglos MN, McCaskill CC et al. Metabolic and behavioral effects of a high-sucrose diet during weight loss. Am J Clin Nutr 1997; 65(4):908-915.
144. Saris WH, Astrup A, Prentice AM et al. Randomized controlled trial of changes in dietary carbohydrate/fat ratio and simple vs complex carbohydrates on body weight and blood lipids: the CARMEN study. The Carbohydrate Ratio Management in European National diets. Int J Obes Relat Metab Disord 2000; 24(10):1310-1318.
145. Yang Q1, Zhang Z1, Gregg EW2, Flanders WD3, Merritt R1, Hu FB4. Added sugar intake and cardiovascular diseases mortality among US adults. JAMA Intern Med. 2014 Apr;174(4):516-24. Doi: 10.1001/jamainternmed.2013.13563.
146. Wolever TM, Jenkins DJ, Jenkins AL, Josse RG. The glycemic index: methodology and clinical implications. Am J Clin Nutr 1991; 54(5):846-854.
147. Thomas DE, Elliott EJ, Baur L. Low glycaemic index or low glycaemic load diets for overweight and obesity. Cochrane Database Syst Rev 2007;(3):CD005105.
148. Radulian G, Rusu E, Dragomir A, Posea M. Metabolic effects of low glycaemic index diets. Nutr J 2009; 8:5.
149. Thomas D, Elliott EJ. Low glycaemic index, or low glycaemic load, diets for diabetes mellitus. Cochrane Database Syst Rev 2009;(1):CD006296.
150. Brand-Miller JC, Holt SH, Pawlak DB, McMillan J. Glycemic index and obesity. Am J Clin Nutr 2002; 76(1):281S-285S.
151. Pi-Sunyer FX. Glycemic index and disease. Am J Clin Nutr 2002; 76(1):290S-298S.
152. Roberts SB. High-glycemic index foods, hunger, and obesity: is there a connection? Nutr Rev 2000; 58(6):163-169.
153. Lindeberg S. Paleolithic diets as a model for prevention and treatment of Western disease. Am J Hum Biol. 2012 Mar-Apr;24(2):110-5. doi: 10.1002/ajhb.22218. Epub 2012 Jan 19.

154. Jönsson T, Granfeldt Y, Ahrén B, Branell U-C, Pålsson G, Hansson A, Söderström M, Lindeberg S. Beneficial effects of a Paleolithic diet on cardiovascular risk factors in type 2 diabetes: a randomized cross-over pilot study. Cardiovascular Diabetology 2009; 8:35-48. Jul 16; doi: 10.1186/1475-2840-8-35.

155. Frassetto LA, Schloetter M, Mietus-Synder M, Morris RC Jr, Sebastian A. Metabolic and physiologic improvements from consuming a paleolithic, hunter-gatherer type diet. Eur J Clin Nutr. 2009 Aug;63(8):947-55. doi: 10.1038/ejcn.2009.4.

156. Martin CA, Akers J. Paleo Diet Versus Modified Paleo Diet: A Randomized Control Trial of Weight Loss and Biochemical Benefit. Journal of the Acadmey of Nutrition and Dietetics 2013; 113 (S3) 9: A-35.

157. Österdahl M, Kocturk T, Koochek A, Wändell PE. Effects of a short-term intervention with a paleolithic diet in healthy volunteers. Eur J Clin Nutr. 2008 May;62(5):682-5.

158. Rizzoli R. Dairy products, yogurts, and bone health. Am J Clin Nutr. 2014 May;99(5 Suppl):1256S-62S. doi: 10.3945/ajcn.113.073056.

159. Turner BL1, Thompson AL. Beyond the Paleolithic prescription: incorporating diversity and flexibility in the study of human diet evolution. Nutr Rev. 2013 Aug;71(8):501-10. doi: 10.1111/nure.12039.

160. Venn BJ1, Mann JI. Cereal grains, legumes and diabetes. Eur J Clin Nutr. 2004 Nov;58(11):1443-61.
161. Forshee RA, Storey ML, Allison DB et al. A critical examination of the evidence relating high fructose corn syrup and weight gain. Crit Rev Food Sci Nutr 2007; 47(6):561-582.
162. Bray GA, Nielsen SJ, Popkin BM. Consumption of high-fructose corn syrup in beverages may play a role in the epidemic of obesity. Am J Clin Nutr 2004; 79(4):537-543.
163. White JS. Straight talk about high-fructose corn syrup: what it is and what it ain't. Am J Clin Nutr 2008; 88(6):1716S-1721S.
164. Melanson KJ, Angelopoulos TJ, Nguyen V, Zukley L, Lowndes J, Rippe JM. High-fructose corn syrup, energy intake, and appetite regulation. Am J Clin Nutr 2008; 88(6):1738S-1744S.
165. Song WO, Wang Y, Chung CE, Song B, Lee W, Chun OK. Ishttp://0-www.ncbi.nlm.nih.gov.helin.uri.edu/pubmed?term=Sergeev IN%5Bauthor%5D&cauthor=true&cauthor_uid=22588363 obesity development associated with dietary sugar intake in the US?. Nutrition http://0-www.ncbi.nlm.nih.gov.helin.uri.edu/pubmed/225883632012; 28:1137-1141.
166. Fulgoni V, III. High-fructose corn syrup: everything you wanted to know, but were afraid to ask. Am J Clin Nutr 2008; 88(6):1715S.
167. White JS. Misconceptions about high-fructose corn syrup: is it uniquely responsible for obesity, reactive dicarbonyl compounds, and advanced glycation endproducts? J Nutr 2009; 139(6):1219S-1227S.
168. Kleiner SM. Water: an essential but overlooked nutrient. J Am Diet Assoc 1999; 99(2):200-206.
169. Otten JJ, Hellwig JP, Meyers LD. Dietary Reference Intakes-DRI: The Essential Guide to Nutrient Requirements. 1st ed. Washington D.C.: National Academies Press, 2006.
170. Lanou A, Barnard N. Dairy and weight loss hypothesis: an evaluation of the clinical trials. Nutrition Reviews 2008; 66(5):272-279.
171. Yanovski J, Parikh S, Yanoff L et al. Effects of calcium supplementation on body weight and adiposity in overweight and obese adults: a randomized trial. Annals of Internal Medicine 2009; 150(12):821-829.
172. Holecki M, Zahorska-Markiewicz B, Wiecek A, Mizia-Stec K, Nieszporek T, Zak-Golab A. Influence of Calcium and Vitamin D Supplementation on Weight and Fat Loss in Obese Women. Obesity Facts: The European Journal of Obesity 2008; 1(5):274-279.
173. Wagner G, Kindrick S, Hertzler S, DiSilvestro R. Effects of various forms of calcium on body weight and bone turnover markers in women participating in a weight loss program. Journal of the American College of Nutrition 2007; 26(5):456-461.
174. Major GC, Alarie F, Dore J, Phouttama S, Tremblay A. Supplementation with calcium + vitamin D enhances the beneficial effect of weight loss on plasma lipid and lipoprotein concentrations. Am J Clin Nutr 2007; 85(1):54-59.
175. Bailey RL, Gahche JJ, Miller PE, Thomas PR, Dwyer JT. Why US Adults Use Dietary Supplements JAMA Int Medicine 2013; 173(5): 355-361
176. 2008 Supplement Business Report. Nutrition Business Journal 2008.
177. Bent S, Tiedt T, Odden M, Shlipak M. The relative safety of ephedra compared with other herbal products. Annals of Internal Medicine 2003; 138:468-471.
178. Dwyer JT, Allison DB, Coates PM. Dietary supplements in weight reduction. J Am Diet Assoc 2005; 105(5 Suppl 1):S80-S86.
179. Greenway FL, de JL, Blanchard D, Frisard M, Smith SR. Effect of a dietary herbal supplement containing caffeine and ephedra on weight, metabolic rate, and body composition. Obes Res 2004; 12(7):1152-1157.
180. Iqbal SI, Helge JW, Heitmann BL. Do energy density and dietary fiber influence subsequent 5-year weight changes in adult men and women? Obesity (Silver Spring) 2006; 14(1):106-114.
181. Howarth NC, Saltzman E, Roberts SB. Dietary fiber and weight regulation. Nutr Rev 2001; 59(5):129-139.
182. Raben A, Jensen ND, Marckmann P, Sandstrom B, Astrup A. Spontaneous weight loss during 11 weeks' ad libitum intake of a low fat/high fiber diet in young, normal weight subjects. Int J Obes Relat Metab Disord 1995; 19(12):916-923.
183. Slavin J. Why whole grains are protective: biological mechanisms. Proc Nutr Soc 2003; 62(1):129-134.
184. Dennis E, Flack K, Davy B. Beverage consumption and adult weight management: A review. Eating Behaviors 2009; 10(4):237-246.
185. Guthrie J, Morton J. Food sources of added sweeteners in the diets of Americans. Journal of the American Dietetic Association 2000; 100(1):43-51.
186. The Beverage Guidance Panel. Beverage Intake in the United States: Noncalorically Sweetened Beverages. 2006. The University of North Carolina at Chapel Hill.
187. Apovian C. Sugar-sweetened soft drinks, obesity, and type 2 diabetes. The Journal of the American Medical Association 2004; 292(8):978-979.
188. Westerterp-Plantenga M, Verwegen C. Short-term effects of an alcohol, fat, protein or carbohydrate preload on energy intake. International Journal of Obesity 1997;(21):S79.
189. Poppitt SD, Eckhardt JW, McGonagle J, Murgatroyd PR, Prentice AM. Short-term effects of alcohol consumption on appetite and energy intake. Physiol Behav 1996; 60(4):1063-1070.
190. Tremblay A, Wouters E, Wenker M, St-Pierre S, Bouchard C, Despres JP. Alcohol and a high-fat diet: a combination favoring overfeeding. Am J Clin Nutr 1995; 62(3):639-644.
191. Duffey K, Popkin B. Adults with Healthier Dietary Patterns Have Healthier Beverage Patterns. The Journal of Nutrition 2006; 136(11):2901-2907.
192. Swithers S, Davidson T. A role for sweet taste: Calorie predictive relations in energy regulation by rats. Behavioral Neuroscience 2008; 122(1):161-173.
193. BioVittoria. Fruit-SweetnessI. http://www.biovittoria.com/Live/biovittoria_1_16.php . 2010.
194. Phelan S, Lang W, Jordan D, Wing RR. Use of artificial sweeteners and fat-modified foods in weight loss maintainers and always-normal weight individuals. Int J Obes (Lond) 2009; 33(10):1183-90. Doi:10.1038/ijo.2009.147.

195. Anderson GH, Foreyt J, Sigman-Grant M, Allison DB. The use of low-calorie sweeteners by adults: impact on weight management. J Nutr 2012; 142(6):1163S-9S. doi:10.3945/jn.111.149617.
196. Womble L, Wang SS, Wadden TA. Commercial and Self-help Weight Loss Programs. In: Wadden TA, Stunkard A, editors. Handbook of Obesity Treatment. London: Guilford Press, 2002: 395-415.
197. Kendall A, Levitsky DA, Strupp BJ, Lissner L. Weight loss on a low-fat diet: consequence of the imprecision of the control of food intake in humans. Am J Clin Nutr 1991; 53(5):1124-1129.
198. Wadden TA, Berkowitz RI, Sarwer DB, Prus-Wisniewski R, Steinberg C. Benefits of lifestyle modification in the pharmacologic treatment of obesity: a randomized trial. Arch Intern Med 2001; 161(2):218-227.
199. Raynor HA, Looney SM, Steeves EA, Spence M, Gorin AA. The effects of an energy density prescription on diet quality and weight loss: a pilot randomized controlled trial. J Acad Nutr Diet 2012; 112(9):1397-402. Doi:10.1016/j.jand.2012.02.020.

200. Pérez-Escamilla R, Obbagy JE, Altman JM, Essery EV, McGrane MM, Wong YP, Spahn JM, Williams CL. Dietary energy density and body weight in adults and children: a systematic review. J Acad Nutr Diet 2012; 112(5):671-84. Doi:10.1016/j.jand.2012.01.020.

201. Raynor HA, Van Walleghen EL, Bachman JL, Looney SM, Phelan S, Wing RR. Dietary energy density and successful weight loss maintenance. Eat Behav 2011; 12(2):119-25. Doi:10.1016/j.eatbeh.2011.01.008.
202.M. Marketdata Enterprises I. The U.S. Weight Loss & Diet Control Market (12th Edition). 12, 1-420. 2013.
203.Academy of Nutrition and Dietetics. Careers in Dietetics: Becoming a Registered Dietitian or Registered Dietitian Nutritionist. 2013.
204. Bureau of Labor Statistics. Occupational Outlook Handbook: Dietitians and Nutritionists. United States Department of Labor. 8 January, 2014.
205. Heshka S, Greenway F, Anderson JW et al. Self-help weight loss versus a structured commercial program after 26 weeks: a randomized controlled study. Am J Med 2000; 109(4):282-287.
206. Summaries for patients. Evaluation of the major commercial weight loss programs. Ann Intern Med 2005; 142(1):I42.
207. Ditschuneit HH, Flechtner-Mors M, Johnson TD, Adler G. Metabolic and weight-loss effects of a long-term dietary intervention in obese patients. Am J Clin Nutr 1999; 69(2):198-204.
208. Brownell KD. The Learn Program for Weight Management. American Health Publishing Company, 2004.
209. Rolls BJ, Barnett R. The Volumetrics Weight Control Plan. New York: HarperCollins Publishers Inc., 2000.
210. Augustin J. Web based resources for weight loss. Nutrition in Clinical Care 2001; 4:272-274.
211. Free Internet Tools for Tracking Weight, Diet, Exercise, and More. FitDay . 2009. 3-3-2007.
212. McCoy MR, Couch D, Duncan ND, Lynch GS. Evaluating an internet weight loss program for diabetes prevention. Health Promot Int 2005; 20(3):221-228.
213. Harvey-Berino J, Pintauro SJ, Gold EC. The feasibility of using Internet support for the maintenance of weight loss. Behav Modif 2002; 26(1):103-116.
214. Bowman SA, Spence JT. A comparison of low-carbohydrate vs. high-carbohydrate diets: energy restriction, nutrient quality and correlation to body mass index. J Am Coll Nutr 2002; 21(3):268-274.
215. Bell EA, Rolls BJ. Energy density of foods affects energy intake across multiple levels of fat content in lean and obese women. Am J Clin Nutr 2001; 73(6):1010-1018.
216. Rolls BJ, Bell EA, Castellanos VH, Chow M, Pelkman CL, Thorwart ML. Energy density but not fat content of foods affected energy intake in lean and obese women. Am J Clin Nutr 1999; 69(5):863-871.
217. Yao MF, Roberts SB. Dietary energy density and weight regulation.(0029-6643 (Print)).
218. Hill JO, Melanson EL, Wyatt HT. Dietary fat intake and regulation of energy balance: implications for obesity. J Nutr 2000; 130(2S Suppl):284S-288S.
219. Boyar AP, Rose DP, Loughridge JR et al. Response to a diet low in total fat in women with postmenopausal breast cancer: a pilot study. Nutr Cancer 1988; 11(2):93-99.
220. Siggaard R, Raben A, Astrup A. Weight loss during 12 week's ad libitum carbohydrate-rich diet in overweight and normal-weight subjects at a Danish work site. Obes Res 1996; 4(4):347-356.
221. Yu-Poth S, Zhao G, Etherton T, Naglak M, Jonnalagadda S, Kris-Etherton PM. Effects of the National Cholesterol Education Program's Step I and Step II dietary intervention programs on cardiovascular disease risk factors: a meta-analysis. Am J Clin Nutr 1999; 69(4):632-646.
222. Prewitt TE, Schmeisser D, Bowen PE et al. Changes in body weight, body composition, and energy intake in women fed high- and low-fat diets. Am J Clin Nutr 1991; 54(2):304-310.
223. Klem ML, Wing RR, McGuire MT, Seagle HM, Hill JO. A descriptive study of individuals successful at long-term maintenance of substantial weight loss. Am J Clin Nutr 1997; 66(2):239-246.
224. Shick SM, Wing RR, Klem ML, McGuire MT, Hill JO, Seagle H. Persons successful at long-term weight loss and maintenance continue to consume a low-energy, low-fat diet. J Am Diet Assoc 1998; 98(4):408-413.
225. McGuire MT, Wing RR, Hill JO. The prevalence of weight loss maintenance among American adults. Int J Obes Relat Metab Disord 1999; 23(12):1314-1319.
226. McGuire MT, Wing RR, Klem ML, Hill JO. Behavioral strategies of individuals who have maintained long-term weight losses. Obes Res 1999; 7(4):334-341.
227. McGuire MT, Wing RR, Klem ML, Seagle HM, Hill JO. Long-term maintenance of weight loss: do people who lose weight through various weight loss methods use different behaviors to maintain their weight? Int J Obes Relat Metab Disord 1998; 22(6):572-577.
228. Polivy J. Psychological consequences of food restriction. J Am Diet Assoc 1996; 96(6):589-592.
229. Hill JO, Thompson H, Wyatt H. Weight maintenance: what's missing? J Am Diet Assoc 2005; 105(5 Suppl 1):S63-S66.
230. The US NEws and World Report. Best Diets. 2014. Available at: http://health.usnews.com/best-diet.
231. Moreno M. The 17 Day Diet. New York. Free Press, 2011.
232. Forberg C, Roberson M, Wheeler L. The Biggest Loser: 6 Weeks to a Healthier You. New York. Rodale Books, 2010.
233. The Mayo Clinic. The Mayo Clinic Diet. New York. Good Books, 2013.
234. Esselstyn R, The Engine 2 Diet. New York. New York. Grand Central Life & Style, 2009.
235. Hand B and Romine S. The Spark Solution. HarperOne, 2014.
236. Asprey D. The Bulletproof Diet. New York. Rodale Books, 2014.
237. Allen JK, Stephens J, Himmelfarb CRD, Sterwart KJ, & Hauck S. Randomized controlled piolet study testing use of smartphone technology for obesity treatment. Journal of Obesity 2013. 23:1-7.
238. Wolfe BM and Belle SH. Long-term risks and benefits of bariatric surgery: A research challenge. JAMA: online, October 1, 2014.
239. Constantine RS, Davis KE, and Kenkel KM. The Effect of Massive Weight Loss Status, Amount of Weight Loss, and Method of Weight Loss on Body Contouring Outcome. Aesthetic Surgery Journal 2014. 34(4): 578-583.
240. Bhaskaran K, Douglas I, Forbes H, dos-Santos-Silva I, Leon DA, and Smeeth L. Body-mass index of 22 specific cancers: a population-based cohort study of 5.2 million UK adults. the Lancet 2014. 384(9945):755-765.
241, Helen W. Lane, PhD, RD (chair); Naomi Trostler, PhD, RD; James O Hill, PHD. Position of the American Dietetics Association: Weight Management. Journal of the American Dietetic Associaiton 2008. 11(41): 330-346.
242. CCC. Calorie Control Council. (2011). 
Sweeteners and lite. Retrieved November 30, 2014, from http://www.caloriecontrol.org/sweeteners-and-lite
243. International Food Information Council Foundation. (2009). 
The lowdown on low-calorie sweeteners . Updated May 23, 2014, from http://www.foodinsight.org/Resources/Detail.aspx?topic=The_Lowdown_on_Low_Calorie_Sweeteners_CPE_Program
244. Fitch C, Kein KS. (2012). Position of the Academy of Nutrition and Dietetics: Use of nutritive and nonnutritive sweeteners. Journal of the Academy of Nutrition and Dietetics, 112, 739-758.
245. Maguire A. (2006). Dental health. In Helen Lucy Mitchell (Ed.), Sweeteners and sugar alternatives in food technology (pp. 19-31). Oxford, UK: Blackwell Publishing Ltd.
246. International Food Information Council Foundation. (2009). Low-calorie sweeteners and health. Updated June 26, 2014, from http://www.foodinsight.org/Resources/Detail.aspx?topic=IFIC_Review_Low_Calorie_Sweeteners_and_Health_
247. Kroger M, Meister K, & Kava R. Low-calorie sweeteners and other sugar substitutes: A review of the safety issues. Comprehensive Reviews in Food Science and Food Safety. 5(2), 35-47.
248. International Food Information Council Foundation. (2004). Sugar alcohols fact sheet. Retrieved October/3, 2011, from http://www.foodinsight.org/Resources/Detail.aspx?topic=Sugar_Alcohols_Fact_Sheet
249. Y.H Hui. (1991). Sweeteners, nonnutritive. Encyclopedia of food science and technology (1st ed., pp. 2470-2487). Australia: Wiley-Interscience.
250. Makinen, K. K. (2011). Sugar alcohol sweeteners as alternatives to sugar with special consideration of xylitol. Medical Principles and Practice : International Journal of the Kuwait University, Health Science Centre, 20(4), 303-320.
251. International Food Information Council Foundation. (2014). FACTS ABOUT LOW-CALORIE SWEETENERS. Retrieved November 30, 2014, from http://www.foodinsight.org/sites/default/files/Facts%20about%20Low%20Calorie%20Sweeteners.pdf
252. Mortensen, A. (2006). Sweeteners permitted in the European Union: safety aspects. Food & Nutrition Research, 50(3), 104-116. doi:10.3402/fnr.v50i3.1588
253. Sandrou, D. K., & Arvanitoyannis, I. S. (2000). Low-fat/calorie foods: Current state and perspectives. Critical Reviews in Food Science and Nutrition, 40(5), 427-447.
254. Ellwein, L. B., & Cohen, S. M. (1990). The health risks of saccharin revisited. Critical Reviews in Toxicology, 20(5), 311-326.
255. The Coca Cola Company: Beverage Institute for Health and Wellness. (2014). Acesulfame Potassium, (Acesulfame K). Retrieved November 30, 2014, from http://www.beverageinstitute.org/en_US/pages/article-potassium.html
256. Magnuson, B. A., Burdock, G. A., Doull, J., Kroes, R. M., Marsh, G. M., Pariza, M. W., et al. (2007). Aspartame: A safety evaluation based on current use levels, regulations, and toxicological and epidemiological studies. Critical Reviews in Toxicology, 37(8), 629-727.
257. The Coca Cola Company: Beverage Institute for Health and Wellness. (2014).
Aspartame safety. Retrieved November 30, 2014, from http://www.beverageinstitute.org/article/aspartame-safety-adi-metabolism-estimated-intakes-and-common-concerns/
258. Whitehouse, C. R., Boullata, J., & McCauley, L. A. (2008). The potential toxicity of artificial sweeteners. AAOHN Journal : Official Journal of the American Association of Occupational Health Nurses, 56(6), 251-9; quiz 260-1.
259. The National cancer Institute. (2009). 
Artificial sweeteners and cancer. Retrieved November/20, 2011, from http://www.cancer.gov/cancertopics/factsheet/Risk/artificial-sweeteners
260. European Food Safety Authority. (2013). Aspartame. Retrieved October 31, 2014, from http://www.efsa.europa.eu/en/topics/topic/aspartame.htm
261. Goyal, S. K., Samsher, & Goyal, R. K. (2010). Stevia (stevia rebaudiana) a bio-sweetener: A review. International Journal of Food Sciences and Nutrition, 61(1), 1-10.
262. Brahmachari, G., Mandal, L. C., Roy, R., Mondal, S., & Brahmachari, A. K. (2011). Stevioside and related compounds - molecules of pharmaceutical promise: A critical overview. Archiv Der Pharmazie, 344(1), 5-19.
263. Ulbricht, C., Isaac, R., Milkin, T., Poole, E. A., Rusie, E., Grimes Serrano, J. M., et al. (2010). An evidence-based systematic review of stevia by the natural standard research collaboration. Cardiovascular & Hematological Agents in Medicinal Chemistry, 8(2), 113-127.
264. Livesey, G. (2001). Tolerance of low-digestible carbohydrates: A general view. The British Journal of Nutrition, 85 Suppl 1, S7-16.
265. Livesey, G. (2003). Health potential of polyols as sugar replacers, with emphasis on low glycaemic properties. Nutrition Research Reviews, 16(2), 163-191.
266. Rangan, C., & Barceloux, D. G. (2009). Food additives and sensitivities. Disease-a-Month : DM, 55(5), 292-311.
267. Blackburn, G. L., Kanders, B. S., Lavin, P. T., Keller, S. D., & Whatley, J. (1997). The effect of aspartame as part of a multidisciplinary weight-control program on short- and long-term control of body weight. The American Journal of Clinical Nutrition, 65(2), 409-418.
268. Brown, R. J., de Banate, M. A., & Rother, K. I. (2010). Artificial sweeteners: A systematic review of metabolic effects in youth. International Journal of Pediatric Obesity : IJPO : An Official Journal of the International Association for the Study of Obesity, 5(4), 305-312.
269. De La Hunty A, Gibson S, & Ashwell M. (2006) A review of the effectiveness of aspartame in helping with weight control. Nutrition Bulletin. 31(2), 115-128.
270. Stowell, J., (2006). Calorie control and weight management. In Helen Lucy Mitchell (Ed.), Sweeteners and sugar alternatives in food technology (pp. 54-61)
271. Rodearmel S.J., Wyatt H.R., Stroebele N., Smith S.M., Ogden L.G., & Hill J.O. (2007). Small Changes in Dietary Sugar and Physical Activity as an Approach to Preventing Excessive Weight Gain: The America on the Move Family Study. Pediatrics. 120, e869
272. Anton, S. D., Martin, C. K., Han, H., Coulon, S., Cefalu, W. T., Geiselman, P., et al. (2010). Effects of stevia, aspartame, and sucrose on food intake, satiety, and postprandial glucose and insulin levels. Appetite, 55(1), 37-43.
273. Sigman-Grant MJ, & Hsieh G. (2005). Reported use of reduced-sugar foods and beverages reflect high-quality diets. The Journal of Food Science. 70(1), S42-S46.
274. American Diabetes Association. (2014). Artificial Sweeteners. Retrieved November 30, 2014, from http://www.diabetes.org/food-and-fitness/food/what-can-i-eat/understanding-carbohydrates/artificial-sweeteners/
275. European Food Safety Authority. (2011). Scientific opinion on the substantiation of health claims related to intense sweeteners and contribution to the maintenance or achievement of a normal body weight (ID 1136, 1444, 4299), reduction of post-prandial glycaemic responses (ID 4298), maintenance of normal blood glucose concentrations (ID 1221, 4298), and maintenance of tooth mineralisation by decreasing tooth demineralisation (ID 1134, 1167, 1283) pursuant to article 13(1) of regulation (EC) no 1924/2006. EFSA Journal, 9(6), 2229. doi:10.2903/j.efsa.2011.2229
276. Tandel, K. R. (2011). Sugar substitutes: Health controversy over perceived benefits. Journal of Pharmacology & Pharmacotherapeutics, 2(4), 236-243.
277. Burt, B. A. (2006). The use of sorbitol- and xylitol-sweetened chewing gum in caries control. Journal of the American Dental Association (1939), 137(2), 190-196.
278. Blundell, J. E., & Hill, A. J. (1986). Paradoxical effects of an intense sweetener (aspartame) on appetite. Lancet, 1(8489), 1092-1093.
279. Rogers, P. J., Carlyle, J. A., Hill, A. J., & Blundell, J. E. (1988). Uncoupling sweet taste and calories: Comparison of the effects of glucose and three intense sweeteners on hunger and food intake. Physiology & Behavior, 43(5), 547-552.
280. Mattes, R. D., & Popkin, B. M. (2009). Nonnutritive sweetener consumption in humans: Effects on appetite and food intake and their putative mechanisms. The American Journal of Clinical Nutrition, 89(1), 1-14.
281. Yang, Q. (2010). Gain weight by "going diet?" artificial sweeteners and the neurobiology of sugar cravings: Neuroscience 2010. The Yale Journal of Biology and Medicine, 83(2), 101-108.
282. Swithers, S. E., Martin, A. A., & Davidson, T. L. (2010). High-intensity sweeteners and energy balance. Physiology & Behavior, 100(1), 55-62.
283. Bellisle, F., & Drewnowski, A. (2007). Intense sweeteners, energy intake and the control of body weight. European Journal of Clinical Nutrition, 61(6), 691-700.
284. Blum J.W., Jacobson D.J., Donnelly J.E. (2005). Beverage consumption patterns in elementary school aged children across two-year period. J Am Coll Nutr, 24(2),93-8
285. Rolls, B.J. (2009).The relationship between dietary energy density and energy intake. Physiology & Behavior, 97:609-615.
286. Pepino, M. Y., & Bourne, C. (2011). Non-nutritive sweeteners, energy balance, and glucose homeostasis. Current Opinion in Clinical Nutrition and Metabolic Care, 14(4), 391-395.

287. Renwick, A. G., & Molinary, S. V. (2010). Sweet-taste receptors, low-energy sweeteners, glucose absorption and insulin release. The British Journal of Nutrition, 104(10), 1415-1420.
288. Stallings, V.A., & Taylor, C.L. (2008). 
Nutrition standards and meal requirements for national school lunch and breakfast programs: Phase I. proposed approach for recommending revisions. Washington, DC: THE NATIONAL ACADEMIES PRESS. Retrieved from http://www.iom.edu/Reports/2008/Nutrition-Standards-and-Meal-Requirements-for-National-School-Lunch-and-Breakfast-Programs-Phase-I-Proposed-Approach-for-Recommending-Revisions.aspx
289. Brown R.J., Walter M., Rother K.I. (2009). Ingestion of diet soda before a glucose oad augments glucagon-like peptide-1. Diab care. 32(12), 2184-6.
290. Weihrauch, M. R., & Diehl, V. (2004). Artificial sweeteners--do they bear a carcinogenic risk? Annals of Oncology : Official Journal of the European Society for Medical Oncology / ESMO, 15(10), 1460-1465.
291. Grabitske, H. A., & Slavin, J. L. (2009). Gastrointestinal effects of low-digestible carbohydrates. Critical Reviews in Food Science and Nutrition, 49(4), 327-360.
292. Seger JC, Horn DB, Westman EC, Lindquist R, Scinta W, Richardson LA, Primack C, Bryman DA, McCarthy W, Hendricks E, Sabowitz BN, Schmidt SL, Bays HE. Obesity Algorithm®, presented by the American Society of Bariatric Physicians®. www.obesityalgorithm.org. (Access = November 2014)

293. Institue of Medicine. Dietary Reference Intakes for Calcium, Phosphorous, Magnesium, Vitamin D, and Fluoride (1997); Dietary Reference Intakes for Thiamin, Riboflavin, Niacin, Vitamin B6, Folate, Vitamin B12, Pantothenic Acid, Biotin, and Choline (1998); Dietary Reference Intakes for Vitamin C, Vitamine E, Selenium, and Carotenoids (2000); Dietary Reference Intakes for Vitamin A, Vitamin K, Arsenic, Boron, Chromium, Copper, Iodine, Iron, Manganese, Molybdenum, Nickel, Silicon, Vanadium, and Zinc (2001); and Dietary Reference Intakes for Calcium and Vitamin D (2011)

294. National Institutes of Health. Office of Dietary Supplements. Dietary Supplement for Weight Loss: Fact Sheet for Health Professionals. October 28, 2014. Available at: http://ods.od.nih.gov/factsheets/WeightLoss-HealthProfessional/

295. CCC. The Calorie Control Council. Aspartame. 2014. Available at: http://www.caloriecontrol.org/sweeteners-and-lite/sugar-substitutes/advantame

296. EFSA Panel on Food Additives and Nutrients Sources Added to Food(2010). Scientific opinion on the safety of steviol glycosides for the proposed uses as food additives. EFSA Journal, 8(4), 1537 – 1621.

297. CCC. The Calorie Control Council. Polyols. 2014. Available at: http://www.caloriecontrol.org/sweeteners-and-lite/polyol.

298. The Obesity Society. Reduced Consumption of Sugar-Sweetened Beverages Can Reduce Total Caloric Intake. April 2014. Accessed November 14,2014 from http://www.obesity.org/publications/reduced-consumption-of-sugar-sweetened-beverages-can-reduce-total-caloric-intake.htm?qh=YToxMTp7aTowO3M6NToic3VnYXIiO2k6MTtzOjY6InN1Z2FycyI7aToyO3M6Nzoic3VnY XJlZCI7aTozO3M6OToic3dlZXRlbmVkIjtpOjQ7czoxMDoic3dlZXRlbmVycyI7aTo1O3M6OToic3dlZXRlbmVyIjtpOjY7czo5OiJiZXZlcmFnZXMiO2k6NztzOjg6ImJldmVyYWdlIjtpOjg7czoxNToic3VnYXIgc3dlZXRlbmVkIjtpOjk7czoyNToic3VnYXIgc3dlZXRlbmVkIGJldmVyYWdlcyI7aToxMDtzOjE5OiJzd2VldGVuZWQgYmV2ZXJhZ2VzIjt9.

299. Peters JC, Wyatt HR, Foster GD, Zhaoxing P, Wojtanowski AC, Vander SS, Herring SJ, Brill C Hill JO. (2014). The effects of water and non-nutritive sweetened beverages on weight loss during a 12-week weight loss treatment program Obesity. 22(247) 1415–1421.

300. Rodearmel S.J., Wyatt H.R., Stroebele N., Smith S.M., Ogden L.G., & Hill J.O. (2007). Small Changes in Dietary Sugar and Physical Activity as an Approach to Preventing Excessive Weight Gain: The America on the Move Family Study. Pediatrics. 120, e869

301. Bryant CE et al Nonnutritive sweeteners: no class effect on the glycemic or appetite responses to ingested glucose 2014 Eur J Clin Nutr doi” 10.10383ejcn 2014.19

302. Espeland MA, Glick, HA Bertoni A et al Impact of an intensive lifestyle intervention on the use and cost of medical services among overweight and obese adults with type 2 diabetes: the Action for Health in Diabetes Diabetes Care 2014;37:2548-2556.

303. Anton SDet al Effects of stevia, aspartame and sucrose on food intake, satiety, and postprandial glucose and insulin levels 2011 Appetite 55: 37-43, Renwick AG and McInarny SV Sweet taste receptors: low energy sweeteners, glucose absorption and insulin release 2010 Brit J Nutrition doi: 10 1017/S0007114519992540

304. Halldorsson TI et al. (2010) Intake of artificially sweetened soft drinks and risk of preterm delivery: a prospective cohort study in 59,334 Danish pregnant women.AJC. 92(3): 626-33

305. USDA. DGAC Meeting 4. 2014. Available at: http://www.health.gov/dietaryguidelines/2015-binder/meeting4/index.aspx.

306. Lutsey PL et al. (2008). Dietary intake and the development of metabolic syndrome. Circulation. 117(6), 754-61.
307. Dhingra R et al. (2007). Soft drink consumption and risk of developing cardiometabolic risk factors and the metabolic syndrome in middle-aged adults in the community. Circulation. 116(5), 480-8

308. Nettleton JA et al, (2009). Diet soda intake and risk of incident metabolic syndrome and type 2 diabetes in the Multi-Ethnic Study of Atherosclerosis (MESA). Diabetes Care. 32(4), 698-94

309. Fung TT et al. (2009). Sweetened beverage consumption and risk of coronary heart disease in women. AJCN. 89(4), 1037-42

310. Duffey KJ et al. (2012). Dietary patterns matter: diet beverages and cardiometabolic risks in the longitudinal Coronary Artery Risk Development in Young Adults (CARDIA) Study. AJCN. 95(4) 909-15

311. Tate DF et al. (2012). Replacing caloric beverages with water or diet beverages for weight loss in adults: main results of the Choose Healthy Options Consciously Everyday (CHOICE) randomized clinical trial. Am J Clin Nutr. 95, 555-563

312. Raben A et al. (2002). Sucrose compared with artificial sweeteners: different effects on ad libitum food intake and body weight after 10 wk of supplementation in overweight subjects. AJCN. 70(4), 721-8

313. Colditz GA et al. (1990). Patterns of weight change and their relation to diet in a cohort of healthy women. Am J Clin Nutr. 51(6), 1100-5

314. Foster SP et al. (2008). Obesity (Sliver Spring).16(8), 1894-900

315. Mazzaffarian D et al. (2011) Changes in Diet and Lifestyle and Long-Term Weight Gain in Women and Men. NEJM. 364(24), 2392- 404

316. de Koning L et al. Sugar-sweetened and artificially sweetened beverage consumption and risk of type 2 diabetes in men. (2011) AJCN. 93(6), 1321-7

317. Miller P and Perez V (2014). Low calorie sweeteners and body weight and composition: a meta-analysis of randomized controlled trials and prospective cohort studies. AJCN. 100, 765-770

318. Phelan S et al. (2009). Use of artificial sweeteners and fat-modified foods in weight loss maintainers and always-normal weight individuals. Int J Obesity (London). 33(10), 1183-90

319. Foreyt J et al. (2012). The Use of Low-Calorie Sweeteners by Children: Implications for Weight Management. J Nutr.142; 155S - 1162S

320. Ebbeling CB et al (2006). Effects of decreasing sugar-sweetened beverage consumption on body weight in adolescents: a randomized, controlled pilot study. Pediatrics.117, 673-80,

321.Williams CL et al. (2007). Weight control among obese adolescents: a pilot study. Int J Food Sci Nutr. 58, 217-30

322. Rodearmal SJ et al. (2007). Small changes in dietary sugar and physical activity as an approach to preventing excessive weight gain: the America on the Move family study. Pediatrics. 120, e869-879

323. Rodearmal SJ et al (2007). Small changes in dietary sugar and physical activity as an approach to preventing excessive weight gain: the America on the Move family study. Pediatrics. 120, e69-79.

324. Ello-Martin JA, Roe LS, Ledikwe JH, Beach AM, Rolls BJ. Dietary energy density in the treatment of obesity: A year-long trial comparing two weight-loss diets. Am J Clin Nutr. 2007 Jun; 85(6): 1,465-1,477.
ADDITIONAL REFERENCES OF INTEREST

Abargouei AS, Janghorbani M, Salehi-Marzijarani M, Esmaillzadeh A. Effect of dairy consumption on weight and body composition in adults: a systematic review and meta-analysis of randomized controlled clinical trials. Int J Obes (Lond) 2012; 36(12):1485-93. Doi:10.1038/ijo.2011.269.

Abete I, Astrup A, Martínez JA, Thorsdottir I, Zulet MA. Obesity and the metabolic syndrome: role of different dietary macronutrient distribution patterns and specific nutritional components on weight loss and maintenance. Nutr Rev 2010; 68(4):214-31. Doi:10.1111/j.1753-4887.2010.00280.x.

Abete I, Parra D, De Morentin BM, Alfredo Martinez J. Effects of two energy-restricted diets differing in the carbohydrate/protein ratio on weight loss and oxidative changes of obese men. Int J Food Sci Nutr 2009; 60 Suppl 3:1-13. Doi:10.1080/09637480802232625.

Acheson KJ. Carbohydrate for weight and metabolic control: where do we stand? Nutrition 2010; 26(2):141-5. Doi:10.1016/j.nut.2009.07.002.

Acheson KJ. Diets for body weight control and health: the potential of changing the macronutrient composition. Eur J Clin Nutr 2013; 67(5):462-6. Doi:10.1038/ejcn.2012.194.

Ajala O, English P, Pinkney J. Systematic review and meta-analysis of different dietary approaches to the management of type 2 diabetes. Am J Clin Nutr 2013; 97(3):505-16. Doi:10.3945/ajcn.112.042457.

Aldrich ND, Reicks MM, Sibley SD, Redmon JB, Thomas W, Raatz SK. Varying protein source and quantity do not significantly improve weight loss, fat loss, or satiety in reduced energy diets among midlife adults. Nutr Res 2011; 31(2):104-12. Doi:10.1016/j.nutres.2011.01.004.

Amato MC, Guarnotta V, Giordano C. Body composition assessment for the definition of cardiometabolic risk. J Endocrinol Invest 2013; 36(7):537-43.

American Dietetic Association. Weight Management. J Am Diet Assoc 2009; 109(2):336-53. Doi:10.1016/j.jada.2008.11.041.

Angelakis E, Merhej V, Raoult D. Related actions of probiotics and antibiotics on gut microbiota and weight modification. Lancet Infect Dis 2013; 13(10):889-99. Doi:10.1016/S1473-3099(13)70179-8.

Arora T, Singh S, Sharma RK. Probiotics: interaction with gut microbiome and antiobesity potential. Nutrition 2013; 29(4):591-6. Doi:10.1016/j.nut.2012.07.017.

Ballesteros-Pomar MD, Calleja-Fernández AR, Vidal-Casariego A, Urioste-Fondo AM, Cano-Rodríguez I. Effectiveness of energy-restricted diets with different protein: carbohydrate ratios: the relationship to insulin sensitivity. Public Health Nutr 2010; 13(12):2119-26. Doi: 10.1017/S1368980009991881.
Bennasar-Veny M, Lopez-Gonzalez AA, Tauler P, Cespedes ML, Vicente-Herrero T, Yañez A, Tomas-Salva M, Aguilo A. Body adiposity index and cardiovascular health risk factors in Caucasians: a comparison with the body mass index and others. PloS one 2013; 8(5):e63999.

Bradley U, Spence M, Courtney CH, McKinley MC, Ennis CN, McCance DR, McEneny J, Bell PM, Young IS, Hunter SJ. Low-fat versus low-carbohydrate weight reduction diets: effects on weight loss, insulin resistance, and cardiovascular risk: a randomized control trial. Diabetes 2009; 58(12):2741-8. Doi:10.2337/db09-0098.

Bray GA. Medical therapy for obesity. Mt Sinai J Med 2010; 77(5):407-417. Doi:10.1002/mjs.20207.

Bray GA, Smith SR, DeJonge L, De Souza R, Rood J, Champagne CM, Laranjo N, Carey V, Obarzanek E, Loria CM, Anton SD, Ryan DH, Greenway FL, Williamson D, Sacks FM. Effect of diet composition on energy expenditure during weight loss: the Pounds Lost Study. Int J Obes (Lond) 2012; 36(3):448-55. Doi:10.1038/ijo.2011.173.

Brooking LA, Williams SM, Mann JI. Effects of macronutrient composition of the diet on body fat in indigenous people at high risk of type 2 diabetes. Diabetes Res Clin Pract 2012; 96(1):40-6. Doi:10.1016/j.diabres.2011.11.021.

Buckley JD, Howe PR. Long-chain omega-3 polyunsaturated fatty acids may be beneficial for reducing obesity-a review. Nutrients 2010; 2(12):1212-30. Doi:10.3390/nu2121212.

Bueno NB, De Melo IS, De Oliveira SL, Da Rocha Ataide T. Very-low-carbohydrate ketogenic diet v. low-fat diet for long-term weight loss: a meta-analysis of

 

Menopause and Hormone Replacement

ABSTRACT

Menopause, defined as twelve months after a woman’s final menstrual period, is a natural event that marks the end of spontaneous ovulation and thus reproductive capabilities. In the Western world, the average age of menopause is 51 years.

During the time preceding and following the menopause, many women experience symptoms including hot flashes, vaginal irritation, trouble sleeping, fatigue, and weight gain. Each woman experiences perimenopause uniquely; although menopause symptoms may represent minor inconveniences for some women, other women find these symptoms more disruptive. This period of a women’s life also coincides with the time she is more likely to develop diseases associated with advancing age such as osteoporosis, cardiovascular disease, and cancer.

The clinical use of estrogens to treat menopausal symptoms was first evaluated in the late 1920s. By 1928, the first commercially available injectable estrogen was developed; and by 1942, the first oral formulation of estrogen was marketed. Over the years, data from clinical studies have refined the indications for hormone therapy. For example, estrogen remains the most effective therapy for hot flashes. However, it has also recently been established that estrogen is not appropriate to prevent chronic disease.

Thus, the challenges to clinicians and patients who consider prescribing and using hormone therapy are: whether to treat; with which agent (formulation, dose, delivery method); and for how long.

THE MENOPAUSAL TRANSITION

Early in the menopausal transition (which starts in the late 40s and lasts about 4 years), ovarian estradiol production is erratic and associated with irregular menstrual cycle
length. FSH levels rise in response to a decrease in levels of inhibin, a protein produced by the granulosa cells (1-7). An FSH level > 10 mIU/ml (measured between cycle day 2-5) indicates ovarian aging. As the final menstrual period approaches, estradiol secretion diminishes and finally ceases. Estrone, derived primarily from peripheral aromatization of androstenedione, becomes the predominant circulating estrogen. The postmenopausal ovary does continue to produce androstenedione and testosterone at premenopausal levels (8). The menopausal transition has been more specifically redefined from the early changes in menstrual cycle length and shortening of the cycle to the full postmenopause(9). However no test at the present time will make the diagnosis and the symptom complex remains the best clinical tool.

A variety of symptoms may accompany the menopausal transition (Table 1). While age at menopause ranges from 49-52 years, cigarette smokers can undergo menopause 1-2 years earlier compared to nonsmokers (10).

THE MENOPAUSAL SYNDROME

Estrogen production during natural menopause does not stop abruptly. For five to seven years before the onset of the last menstrual period, ovarian function begins to diminish. Menopause can also be induced

TABLE 1: selected menopause symptoms
Abnormal uterine bleedingVasomotor symptoms*

Vulvovaginal dryness, irritation, atrophy*

Urinary incontinence

Trouble sleeping*

Sexual dysfunction

Dyspareunia

Depression

Anxiety

Labile mood

Fatigue

Headache

Myalgias

Arthralgias

Weight gain

Poor memory

Dry skin

Dry eyes

Thinning scalp hair

Hirsutism

surgically (i.e. bilateral oophorectomy) or medically (e.g. chemotherapy or pelvic irradiation). Because ovarian estrogen levels fall abruptly with induced menopause, these women generally experience more severe menopausal symptoms (11, 12). Menopause occurring at or before age 40 is called premature menopause; up to 90% of cases of spontaneous premature menopause (primary ovarian insufficiency) are idiopathic.

After the last menstrual period, the ovary ceases to secrete estradiol. However, smaller amounts of a weaker estrogen, estrone, are still synthesized from androstenedione in the cortex of the adrenal gland and in the interstitial ovarian cells (in minor amounts)(8). Small amounts of this estrone can be transformed into estradiol.

Body mass is directly correlated with the rate of peripheral production of estrone and estradiol in postmenopausal women. Estrogen synthesis takes place largely in adipose tissue. Therefore, an obese woman may produce twice as much estrone and estradiol as a thin woman. This may help explain the increased prevalence of hypo-estrogenemic symptoms and the higher risk of osteoporosis observed in thin women.

PIVOTAL STUDIES

The results of the large Women’s Health Initiative (WHI) study have been both influential and controversial. In 2002, the estrogen-progestin arm of the WHI was stopped prematurely because of increases in the risk of breast cancer and coronary heart disease (13). In 2004, the estrogen-only arm was also prematurely discontinued, reporting that estrogen therapy had no effect on CHD risk and increased the risk of stroke and deep vein thrombosis in this population(14). Post-hoc analyses suggest no increase in CHD in women starting treatment within 10 years of menopause(15). Other doses and types of estrogens and progestins were not studied in WHI; smaller studies are now underway to further investigate whether age at therapy initiation and different types/doses of estrogens and progestins will result in different health outcomes. Table 2 provides a brief outline of pivotal studies (including ongoing ones) involving perimenopausal women.

The findings of the Women’s Health Initiative study, a prospective, randomized trial of more than 16,000 healthy, post-menopausal women, published in July 2002, have thrown the use of HT into question in both the medical and lay communities. The estrogen plus progestin arm of the study was halted because there was a small, increased risk of invasive breast cancer among women receiving the combined therapy, as well as an increased risk of heart attacks, stroke and clotting. These risks were not offset by the benefits: a decrease in colon cancer and hip fractures (13).

However, the average age of the women in the WHI was 63.2 years, and does not reflect normal clinical practice where replacement is used mainly for symptoms, including hot flashes, in women 10 to 30 years younger. Women in the WHI also had an average BMI of 28, one-third had hypertension, and one-half had a history of smoking (66). Thus, at the present time, the relative risk to benefit of using HT in younger, healthier women is largely unknown, and physicians cannot make all clinical decisions based on the WHI study, as it appears to apply specifically to the population studied. Hormone replacement is still an important therapeutic modality for women with symptoms and quality of life issues which deserves further study, and should be considered by physicians for their patients on an individual basis. At the present time there is global consensus that women with early or premature menopause should be treated until the normal age of menopause (age 50) and their treatment during these years should not be considered in the calculation of years of postmenopausal therapy(16).

TABLE 2: Important studies involving perimenopausal women

Study

Type

Location

Dates

N

Ages; mean

Hormone formulation

WHI (E+P)

RCT

US

1993-2002

16608

50-79; 63

CE 0.625 mgMPA 2.5 mg
Intact uterus. Stopped early after 5.2 yrs (planned for 8); increased CHD events & invasive breast cancer.
WHI (E)

RCT

US

1993-2004

10739

50-79; 63

CE 0.625 mg
Status-post hysterectomy. Stopped early after 6.8 yrs; increased risk CVA; lack of CHD benefits.
PEPI

RCT

US

1989-1994

875

45-64; 67

CE 0.625 mg± MPA 10 mg (days 1-12)± MPA 2.5 mg QD

± P4 200 mg (days 1-12)

Healthy women; 3 years follow-up. HT improved lipoprotein profiles. Unopposed estrogen associated with high rate endometrial hyperplasia.
HERS

RCT

US

1993-1998

2763

67

CE 0.625 mgMPA 2.5 mg
Subjects had known CHD. HT 36 months.
NHS

obs

34-59

SWAN

obs

US

1996-current

3302

42-52

As per patient preference (including no HT)
Multiracial, multiethnic (Caucasian, African American, Hispanic, Chinese, Japanese); includes premenopausal; yearly visits—currently tracking 12th-13th visits. Following bone density, cardiovascular health, mood, symptoms.
MWS

obs

UK

1996-current

1084110

50-64; 56

As per patient preference
WISDOM

RCT

UK, Australia, New Zealand

1999-2002

5692

50-69; 63

CE 0.625 mg± MPA 2.5 or 5 mg
Stopped early after median 12 months follow-up (planned 10 yrs) because of WHI results.
ELITE

RCT

US

2004-2013

643

6 yrs vs. 10 yrs postmeno.

E2 1 mg PO
2.5 yrs planned; endpoint atherosclerosis by carotid ultrasound.
KEEPS

RCT

US

2005-2012

727

42-58,52

CE 0.45 mgor E2 50 mcg transdermal P4 200 mg (days 1-12)
Harvard Mood

obs

US

1995-2006

460

36-45

No RX
(DOPS)Schierback et al

RCT

Denmark

1990-2008

1006

49.7±2.8

2 mg synthetic 17-β-estradiol for 12 days, 2 mg 17-β-estradiol plus 1 mg norethisterone acetate for 10 days, and 1 mg 17-β-estradiol for six days or 2mg 17β estradiol for hysterectomized

 


Abbreviations:

WHI, Women’s Health Initiative (13, 14).

PEPI, Postmenopausal Estrogen/Progestin Interventions (17).

HERS, Heart and Estrogen/progestin Replacement (18).

NHS, Nurses Health Study(19-22).

SWAN, Study of Women’s Health Across the Nation (23-25).

MWS, Million Women Study (26).

WISDOM, Women’s International Study of long Duration Estrogen after Menopause (27).

ELITE, Early vs. Late Intervention Trial with Estradiol. (28)

KEEPS, Kronos Early Estrogen Prevention Study.

Harvard Study of Moods and Cycles(29)

DOPS Danish Osteoporosis Prevention Study(30)

RCT, randomized controlled trial.

obs, observational study.

CE, conjugated estrogens.

MPA, medroxyprogesterone acetate.

E2, estradiol.

P4, micronized progesterone.

The results of the large Women’s Health Initiative (WHI) study have been both influential and controversial. In 2002, the estrogen-progestin arm of the WHI was stopped prematurely because of increases in the risk of breast cancer and coronary heart disease (13). In 2004, the estrogen-only arm was also prematurely discontinued, reporting that estrogen therapy had no effect on CHD risk and increased the risk of stroke and deep vein thrombosis in this population(14). Post-hoc analyses suggest no increase in CHD in women starting treatment within 10 years of menopause(15). Other doses and types of estrogens and progestins were not studied in WHI; smaller studies are now underway to further investigate whether age at therapy initiation and different types/doses of estrogens and progestins will result in different health outcomes. Table 2 provides a brief outline of pivotal studies (including ongoing ones) involving perimenopausal women.

The findings of the Women’s Health Initiative study, a prospective, randomized trial of more than 16,000 healthy, post-menopausal women, published in July 2002, have thrown the use of HT into question in both the medical and lay communities. The estrogen plus progestin arm of the study was halted because there was a small, increased risk of invasive breast cancer among women receiving the combined therapy, as well as an increased risk of heart attacks, stroke and clotting. These risks were not offset by the benefits: a decrease in colon cancer and hip fractures (13).

However, the average age of the women in the WHI was 63.2 years, and does not reflect normal clinical practice where replacement is used mainly for symptoms, including hot flashes, in women 10 to 30 years younger. Women in the WHI also had an average BMI of 28, one-third had hypertension, and one-half had a history of smoking (66). Thus, at the present time, the relative risk to benefit of using HT in younger, healthier women is largely unknown, and physicians cannot make all clinical decisions based on the WHI study, as it appears to apply specifically to the population studied. Hormone replacement is still an important therapeutic modality for women with symptoms and quality of life issues which deserves further study, and should be considered by physicians for their patients on an individual basis. At the present time there is global consensus that women with early or premature menopause should be treated until the normal age of menopause (age 50) and their treatment during these years should not be considered in the calculation of years of postmenopausal therapy(16).

This article will review the current state of knowledge concerning menopause and menopausal hormone therapy (HT). Unless otherwise noted the term HT will be used in this chapter to refer to use of estrogen and a progestogen in women with a uterus and to use of estrogen alone in women who do not have a uterus. The following questions will be addressed: What is the effect of HT on hot flashes, genitourinary tract atrophy, and other symptoms of the menopausal syndrome? Does HT reduce a woman's risk of osteoporosis, cardiovascular disease, or cancer of the breast, endometrium, or colon? Can HT slow the decline of cognitive function and prevent Alzheimer's disease? What recommendations should women be given in light of the Women’s Health Initiative findings?

VASOMOTOR SYMPTOMS

A hot flash is a transient feeling of warmth especially over the face and neck, which lasts for several minutes. For some women, hot flashes are associated with drenching sweats, increased heart rate, and post-flash chills. When they occur at night, they can cause sleep disturbances, fatigue, and depression. Vasomotor symptoms typically begin during the menopausal transition, reach maximal frequency and intensity during the two years after menopause, and gradually subside—ultimately lasting 1-5 years. Perhaps 75% of perimenopausal women experience hot flashes (5.) Up to 10% of women experience hot flashes for 10 years or longer. Recent data document vasomotor symptoms for a mean duration of 7.4 years with women starting in the pre or perimenopause having a longer duration ( 9.4 to 11.8 years) than women starting in the postmenopause (3-4 years). Prevalence of hot flashes differs by culture and ethnicity and can range from 0-80%. African Americans appear to have the longest duration (10.1 years) followed by Hispanics (8.9 years) followed by non-hispanic whites (6.5 years) Chinese (5.4years) and Japanese the shortest, (4.8 Years) (11, 25, 31-33). Hot flashes occur with greater frequency in women who undergo surgical menopause than in those who experience natural menopause. They are also more common at night, which often results in sleep disturbances, fatigue and depression. Additional factors including warm environments, consumption of alcohol or caffeine, and stress can exacerbate hot flash occurrence.

Several studies have documented the effectiveness of estrogen therapy (in various formulations, doses, and delivery methods) for hot flashes (11, 34). For example, the Women’s Health, Osteoporosis, Progestin, Estrogen (HOPE) study randomized postmenopausal women to 3 doses of oral conjugated equine estrogens (CEE) 0.625 mg/d, 0.45 mg/d, 0.3 mg/d; with or without medroxyprogesterone acetate (MPA) 2.5 mg/d or 1.5 mg/d. Among the 241 subjects, there was a was a significant reduction in the number and severity of hot flashes as compared to baseline and placebo after three weeks of hormone therapy(35). Average daily hot flashes fell from nine per day at baseline to two per day after one year of therapy. This reduction was seen in all doses of CEE and CEE/MPA, including the lowest doses (35). In one comparative 12-week trial of 204 postmenopausal women (average age, 52 years), oral CEE 0.625 mg and transdermal estradiol 50 mg/day provided similar symptom relief (36)

SLEEP DISTURBANCES

HT for relief of hot flashes is effective in up to 90% of menopausal women. However, many women still experience sleep disturbances. Poor sleep (including difficulty falling asleep, disrupted sleep, insufficient quantity, and poor quality) affects approximately 45% of perimenopausal women in the U.S. and is associated with reduced productivity, irritability, depression, and cardiovascular disease (37, 38). The causes are multiple and include: hot flashes, nocturia, anxiety, depression, and primary sleep disorders (i.e. apnea, periodic limb movements/restless legs syndrome).

Time of night (i.e. first vs. second half) appears to influence the etiology of poor sleep. Laboratory sleep studies have shown that hot flashes tend to cause arousals in the first half of the night and are associated with subjective poor sleep. However, apneas and periodic limb movements tend to cause arousals in the second half of the night—a time when rapid eye movement (REM) sleep predominates and suppresses thermoregulatory effector responses like hot flashes. Thus in the second half of the night, primary sleep disorders cause arousals (hence loss of REM sleep) and may subsequently precipitate hot flashes (39, 40).

Determinants of subjective versus objective sleep quality may also be different. Whereas subjective sleep quality tends to be lower in women who experience hot flashes and report anxiety, objective sleep efficiency (ratio of time-asleep to time-spent-in-bed) tends to be lower in women who have apnea or periodic limb movements (40).

Although it is often assumed that complaints of poor sleep are due to hot flashes, treatment of hot flashes may not improve sleep quality if there is an underlying primary sleep disorder or psychiatric condition syndrome. Thus, providers should assess patients for apnea, restless legs, anxiety, and depression, and consider appropriate treatment.

A double-blind, crossover study in hypogonadal postmenopausal women compared the effects of CEE (0.625 mg/d) and placebo on sleep patterns(41). Results showed no relation between hot flashes or night sweats and sleep disturbances. For the women on estrogen, however, sleep quality improved, along with length of REM sleep and sleep latency. The causes of sleep disturbances in the postmenopause are complex and further research is needed.

DEPRESSION

Although prior epidemiologic studies have concluded that postmenopausal women are not at increased risk for depression, studies in the past few years including the Harvard study of Moods and Cycles have shown depressive symptoms are observed more frequently in this during the menopausal transition.(42). Increased psychological distress was seen in the SWAN study of women in the transition (29, 43, 44). This vulnerability occurs even if women do not have a previous history of depression (44, 45), although women with a previous history appear to be at greater risk (46), and depression is more apt to occur in the later stages of perimenopause (47-50). In particular, a subgroup of women who show abnormal mood responses to periods of estrogen withdrawal such as postpartum and premenstrually with a diagnosis of PMDD (premenstrual dysphoric disorder) may be especially vulnerable (51, 52). Women suffering from hot flashes (53) and sleep disorder(54) are also vulnerable, but depressive episodes can occur regardless of the presence of hot flashes(55).Women with early onset of perimenopause also have a significantly increased risk of first onset depression(42, 56) as well as those with a longer length of perimenopause(47, 48). Thus hormonal fluctuations may be a psychological destabilizer and there is some evidence that sex hormones may prevent, attenuate or even treat depressive episodes in the perimenopause (50, 57). This is an area of evolving research, but two independent studies demonstrate successful treatment of depression with transdermal estradiol (49, 58). This treatment; however, does not appear to be successful following menopause (59). Whether asymptomatic postmenopausal women benefit is unproven but the KEEPS study showed improvement in depression, anxiety and sexual function(60) Current recommendations include the use of hormone therapy as well as selective serotonin reuptake inhibitors when treating refractory depression (61). However, the progestin in these patients should be chosen carefully as depressive symptoms may occur with these medications, and some patients cannot tolerate any progestin, even progesterone(62).

With regard to mood, the data supports the theory that symptoms of depression may be alleviated with the use of ET/HT. In a double-blind placebo controlled trial of perimenopausal women, Soares et al found that depressive symptoms were significantly relieved in women receiving estradiol compared to placebo (58). In addition, estrogen has been shown to improve mood in post menopausal women without clinical depression (63, 64). Further research is still needed in this area, but the preliminary data suggest that estrogen may be a possible treatment method for some depression symptoms.

GENITOURINARY TRACT ATROPHY

Large numbers of estrogen receptors are found in the vagina, vulva, urethra, and trigone of the bladder. Thus, atrophy of the genitourinary tract can occur as estrogen levels diminish.

Vulvovaginal atrophy causes significant complaints and is common in the menopause. Symptoms include dryness, dyspareunia, discharge, itching and occasionally bleeding. The symptoms increase with age and may lead to vulvovaginal fissures and stenosis. These symptoms are described as moderate to severe in the majority of women who report them which in one survey was 30%(65).

After menopause, the vaginal walls thin and lose their elasticity. They also produce fewer secretions and lose much of their lubricating ability in response to sexual stimuli. The vulva becomes flattened and thin as a result of the loss of collagen, adipose tissue and the ability to retain water(66). The urethra also becomes thinner and less efficient, with detrusor pressure at the urethral opening decreasing, both during and after voiding. Estrogen deficiency also leads to an increase in fibrosis of the bladder neck, reduced collagen in surrounding tissues, and a decrease in the number and diameter of the muscle fibers in the pelvic floor. There is a decrease in the superficial layer of the vaginal epithelium, a decrease in vaginal secretions and pH (normal is under 4.5) and an increase in vaginal infections due to loss of the normal acidic environment and overgrowth of opportunistic fecal bacteria at the expense of normal lactobacilli. Loss of subcutaneous fat leads shrinkage of the labia and retraction of the urethra(67) Estrogen treatment, both systemic and local can greatly relieve these problems(67-69).

These changes increase a woman's risk of vaginal and urinary tract infection. Atrophic genitourinary tissues are also at increased risk of injury by trauma. Estrogen replacement therapy can significantly lessen these problems. The advantage of using local vaginal therapy is that minimal if any absorption occurs after the first two weeks of therapy, and it can be used without the side effects of systemic therapy. Multiple studies have shown that the absorption of vaginal estrogen therapy is strictly dose dependent and is maximal in the first two weeks of treatment when the vagina is thin and atrophic. With the return of the normal superficial layer of the vagina, serum levels of estradiol remain in the postmenopausal range when used in minimal doses. Studies have followed women for up to 3 months including patients with breast cancer (69-71). Endometrial safety is maintained if the local therapy is minimal (0.5 grams of cream or 10-25ug of the vaginal pill twice a week or the equivalent) but evaluation is warranted for any bleeding(72).

SEXUAL DYSFUNCTION

All of the changes to the genitourinary tract can result in dyspareunia, leading to a decreased interest in sexual intercourse. Fatigue and depression brought on by the vasomotor symptoms and sleep disturbances of menopause can exacerbate this lack of interest in coitus.

Decreased levels of endogenous testosterone, both in women who have undergone surgical menopause, as well as in those who experience natural menopause, may cause decreased libido(73). Women who complain of lack of sex drive may be candidates for androgen replacement, as well as estrogen. In general, androgen levels do not decrease abruptly at menopause but decrease gradually as women age so that decreased libido may be a problem of older postmenopausal women.

OSTEOPOROSIS

The loss of ovarian hormone production after menopause puts women at increased risk for osteoporosis. Without estrogen, osteoclast activity and bone resorption are increased, and bone mass decreases. This reduced skeletal mass and microarchitectural deterioration increase the risk of fracture. At age 50, a Caucasian woman has a 16% lifetime risk of hip fracture, a 15% risk of a Colles’ fracture, and a 32% chance of an atraumatic vertebral fracture (74).

Peak bone mass—typically attained by the third decade of life—is determined by genetic and environmental (nutrition, lifestyle, physical activity) factors (75). There is often only slight bone loss between age 30 and the perimenopausal transition (76). The period of accelerated bone loss appears to last approximately 5 years starting 2 years before the final menstrual period lasting until 2-4 years following the final menstrual period. For example, a prospective study of 75 Caucasian women followed for 9.5 years found that subjects lost 10.5% of bone at the lumbar spine over the critical 5-year period while estrogen levels were declining (77).

Although estrogen therapy can prevent bone loss and reduce the risk of fracture in perimenopausal women, it is no longer recommended as first-line therapy for osteoporosis because of the risks associated with hormone therapy and because alternative therapies exist. Studies have shown estrogen to not only prevent bone loss (by decreasing osteoclastic activity), but also to reduce fracture rates by as much as 65%(12). Women in the Women’s Health Initiative receiving estrogen plus progestin suffered 5 fewer hip fractures per 10,000 compared to women on placebo (66). Estrogen therapy may be considered in women for whom the alternative agents (e.g. bisphosphonates, raloxifene, teriparatide) are intolerable or contraindicated; in whom estrogen therapy is also indicated for other reasons (e.g., vasomotor symptoms); or in whom the benefits of estrogen therapy outweigh the risks (78). In the WHI, women who received estrogen plus progestin had hazard ratios of 0.66 for hip fractures, 0.66 for vertebral fractures, and 0.77 for fragility fractures at any site, as compared to women on placebo (13). Prophylactic benefit increases when estrogen replacement is begun as soon after menopause as possible. Because bone loss continues as soon as estrogen replacement is stopped, treatment will be needed so as to maintain the positive effects on bone metabolism. However, the longer a woman has been taking estrogen, the more bone she will have when treatment is stopped and bone loss resumes.

While estrogen prevents bone loss in most postmenopausal women, some continue to lose bone mass despite the therapy, presumably because of genetic or environmental factors. Bone density studies should be conducted during the perimenopausal period and then repeated as needed to assess the status of bone loss(79).

All postmenopausal women should have an appropriate calcium intake (up to 1200 mg) and vitamin D (400 IU) supplementation. Concerns about calcium supplements and cardiovascular disease from both observational and randomized studies have changed recommendations, although the issue is controversial(80). Calcium is best obtained from food and women should aim to meet requirements primarily through nutrition and take supplements only if needed to reach RDA .Mean dietary intake of midlife and older women is 700mg/day (81) so supplement in the range of 500mg is appropriate when dietary intake of calcium is low. Recent studies have shown that more than 50% of women over age 50 are vitamin D insufficient and these replacement doses are probably inadequate(82). 1000-2000 IU of Vit D3 are probably a better estimate of replacement. Only 10% of calcium is absorbed when Vitamin D is low. Supplements can help compensate for poor dietary intake of calcium and inefficient vitamin D synthesis. Because calcium carbonate requires acid for absorption, women taking acid-suppressing drugs or with atrophic gastritis should take calcium citrate, which does not require gastric acid for absorption.

CARDIOVASCULAR DISEASE

Cardiovascular disease accounted for 30.7% of deaths in American women in 1999. It surpasses cancer, cerebrovascular disease, lung disorders, infectious disease, diabetes, suicide, and renal disease as the leading cause of death in women today(83). A woman has about 10 times the lifetime risk of dying of ischemic heart disease than of breast cancer, reproductive cancer, or osteoporotic fracture.

An acceleration of heart disease occurs after age 50, and approximately one third of the women who die of cardiovascular disease every year are under 65 years old (more than 100,000). This suggests that menopause (whether surgical, premature, or natural) may be a risk factor for heart disease(84). Because premenopausal women have lower incidences of cardiovascular disease than men and lose this advantage after menopause, it is logical to conclude that estrogen has a cardioprotective effect. It is thought that estrogen deficiency is at least partially responsible for the increased risk of developing heart disease after menopause.

Considerable controversy and confusion has recently erupted over the role of estrogen replacement therapy in preventing cardiovascular disease. A number of trials reported an increased risk of ischemic events when hormone therapy was started in older women with a history of heart disease(21, 85, 86). In response, the American Heart Association recommended that hormone replacement therapy not be used for primary prevention of cardiovascular disease. The results of the estrogen-progestin arm of the WHI showed similar results. Women on estrogen-progestin therapy suffered 7 more CHD events per 10,000 women than women on placebo. They also suffered 8 more strokes per 10,000 women than those taking placebo(13).

Emerging evidence suggests hormone therapy is most effective in protecting women whose hearts are not yet compromised from future cardiovascular disease as seen in a recent study by Hodis et al.(87). Researchers randomized 222 postmenopausal women with no history of cardiovascular disease, stroke, or cancer who had high levels of LDL (≥ 130 mg/dL) to receive either 1 mg unopposed 17-ß estradiol or placebo. After two years, women on estrogen had significantly less thickening of the inner carotid artery wall. Recent data published from the WHI study show that the risk of coronary heart disease is largely dependent on age of the women initiating therapy and the number of years since menopause. The lower risk in the 50 to 59 year age group and in those experiencing menopause within the last 10 years (15, 88) and those on therapy more than 6 years (15). Data on estrogen treatment alone in WHI showed a decrease in coronary calcium, particularly in younger women although the effect was observed in all ages (89). In contrast to these findings, other publications from the same study suggested that the gap between menopause and initiation of therapy has no effect on cardiovascular disease, contradicting their previous report which showed some protection if started early (90, 91). However these observations are from a combination of the randomized and observational studies with most women who were recently menopausal were previously taking hormone therapy. One study showed some protection after 6 years of use(92). Overall, most studies have shown convergence between the observational and the randomized control publications suggesting that younger women starting hormone therapy at menopause are not at increased risk for heart attacks (93). The KEEPS study examined the effects of hormone treatment on surrogate markers of cardiovascular disease in recently menopausal women including carotid intima-media thickness (IMT) and coronary calcium. Carotid IMT increased in a similar fashion in both treated and placebo groups and there was a non-significant trend for less coronary calcium in the hormone arms(94) The DOPS study followed women on hormone therapy for 16 years and although osteoporosis was the primary endpoint, mortality and hospitalizations for both congestive heart failure and MI was reduced in the treated arms. Younger women appeared to show more benefit(30) Probably most convincing are the results of the Elite trial showing that younger recently women treated with hormone therapy showed an attenuation of IMT thickness while women treated who were 10 years past menopause showed no such benefit(95). When women stopped therapy in WHI, the increased risk seen in the treated arm was no longer apparent after a mean of 2.4 years (96). Endothelial dysfunction, not atherosclerosis, appears to be significantly increased in women with hot flashes, perhaps explaining their increased cardiovascular risk profile (97). Since symptomatic women were not studied in WHI, the role of HT in relief of symptoms and in turn of their effect on coronary risk is unclear. An

However, at the present time, HT should not be recommended for the prevention of heart disease.

ALTERED LIPOPROTEIN PROFILES

The increased risk of cardiovascular disease after menopause might be explained by the atherogenic changes in plasma lipoprotein levels associated with estrogen deficiency. At menopause, plasma levels of low-density lipoprotein (LDL) increase by 10% to 15%. According to data from the SWAN study (Study of Women’s Health Across the Nation), women experience a very specific increase in lipids at menopause. This includes total cholesterol, low-density lipoprotein cholesterol, and apoliprotein B. These changes were similar across all ethnic groups (98).

This increase can be prevented with estrogen replacement therapy. Plasma levels of high-density lipoprotein (HDL) increase by 10% to 15% with estrogen therapy and may be an important factor in the cardioprotective effect of estrogen.

The use of progestins, however, in conjunction with estrogen seems to attenuate these beneficial effects on plasma lipoprotein levels to some extent. Data from the Nurses' Health Study showed that women who took estrogen and progestin in combination had the same apparent protection from coronary events as did the women who took estrogen alone(21).

However, as noted previously, the randomized HERS trial showed that HT (with 0.625 mg/d of conjugated equine estrogen and 2.5 mg/d of medroxyprogesterone acetate) increased the risk of coronary events in women with a mean age of 65 who had established cardiovascular disease(18). This effect was noted during the first year of HT use. Following the second year, a progressive protective trend was found with HT, although there was no overall beneficial effect in the study as a whole. Another study examined the effect of HT/ERT as well as ERT in women with angiographically verified coronary disease(99). Again, no benefit was seen. However, these women had proven heart disease and were, on average, 65 years of age. This is considerably older than the age when HT is usually started. These data suggest that HT raised the possibility that started prior to the development of cardiovascular disease might be protective.

Progestins may have variable effects on lipoproteins based on their androgenicity. More androgenic progestins tend to lower HDL levels to a greater degree than do the less androgenic progestins (100). The two types of progestins most commonly used for hormone replacement therapy are those derived from 19-norestosterone and 17-hydroxyprogesterone. The former are the more androgenic, while the latter have a little androgenicity. Medroxyprogesterone is the most commonly prescribed progestin in the United States and is derived from 17-hydroxyprogesterone.

More recently, micronized progesterone has become available. The Postmenopausal Estrogen/Progestin Interventions Trial (PEPI) showed that micronized progesterone, used with conjugated equine estrogen, had less attenuation of the favorable lipid profile induced by estrogen than medroxyprogesterone acetate (101).

VASODILATION

As important as estrogen's effects on lipid metabolism may be its vasodilatory properties. It appears to potentiate the effects of endothelium-derived relaxing factor (EDRF) in the coronary arteries. It also may affect vasodilation through an endothelium-independent pathway in the peripheral vasculature.

One study looking at postmenopausal women with angina and normal coronary arteries (syndrome X) saw diminished vasodilation before initiation of estrogen therapy and normalized hyperemic response after two months of treatment. Vasodilation was measured by testing hyperemic response to forearm blood flow occlusion. Chest pain either improved markedly, or resolved, in 19 of the 20 subjects. This improvement in angina symptoms suggests that the impaired vasodilatory response to an EDRF/nitric oxide stimulus may be systemic(102).

An additional study reported a beneficial effect for sublingual estradiol in reducing symptoms of exercise-induced myocardial ischemia in postmenopausal women with coronary artery disease(103). These results suggest both a reduction in peripheral vascular resistance and a direct vasodilatory effect in the coronary arteries.

OTHER EFFECTS

Additional studies have found an association between HT and a marked reduction in the pulsatility index of the internal carotid and middle cerebral arteries(104). This finding may help explain the reduction in stroke risk and the improvement in cognitive function seen with estrogen plus progesterone. According to recent data from the Nurses' Health Study, this effect is seen at low doses only (0.3 mg conjugated equine estrogen).

A recent study also looked at the effects of estrogen in women who had recently suffered ischemic stroke or transient ischemic attacks and found no reduced mortality or recurrence prevention with 1.0 mg estradiol compared to placebo(105). These findings discourage the use of HT for secondary stroke prevention.

Other factors associated with estrogen use which could lower the risk for cardiovascular disease include decreases in levels of the proatherosclerotic factor, lipoprotein (a), the procoagulant factor, fibrinogen, and increases in levels of factor 11 (prothrombin). One study showed that with discontinuation of hormone therapy there was a rise in use of antihypertensive medication(106).

BREAST CANCER

One in 8 women will be diagnosed with breast cancer in her lifetime, and risk increases with age(107). In 2001, approximately 40,200 women died of breast cancer, although survival rates have been increasing. The five-year survival rate for women with localized breast cancer has risen from 72% in the 1940s to 97% today. This high survival rate, however, decreases to 77% if the cancer has spread regionally, and to 21% if it has spread distantly(107).

Estrogen, a trophic growth hormone, may promote the growth of preexisting breast cancer. It is still unknown whether it may also induce the growth of new cancers. Use of estrogen alone for at least five years, may be associated with a slightly increased risk of breast cancer according to the Nurses' Health Study. However, a report from the Women’s Health Initiative study showed an small increase in breast cancer in women on estrogen plus progestin , women on estrogen only showed no increased incidences of breast cancer compared to women on placebo (13, 14). Recent publications showed a significant decrease in the incidence of breast cancer in this group(108), a surprising finding which may be related to the type of estrogen used in the WHI study (conjugated equine estrogen). The study is ongoing but clarification of this discrepancy has not been forthcoming. The relative risk of the Estrogen plus Progestin (E+P) arm of the study has varied from 1.24 to 1.28 and follow up publication from WHI showed a non significant risk of 1.20 (0.94-1.53)(109). It has been suggested that the effect of E+P is to promote the growth of occult tumors which are present on the initiation of therapy.(110)The risk is very small although the data interpretation has implied otherwise. The absolute number of excess cases is stated as 8/10000 per year and is related to cumulative exposure. Women who had never received hormones in the past in WHI did not have a significant risk over the 5.6 years of the trial and the risk was not significant in younger women.(111)There was no increase in risk for at least 7 years(109)

Many studies have not shown an increased risk of breast cancer with estrogen use. A large meta-analysis of 51 epidemiologic studies (involving more than 160,000 women from 21 countries) showed that HT increases the risk of breast cancer and that risk increases with longer use(112). That is, for every 1,000 women who began using HT at age 50 and continued using it for 5, 10, or 15 years, an additional 2, 6, or 12 cases of breast cancer would be expected to occur. However, another review showed that at doses of 0.625 mg/d conjugated estrogens, there was no increased risk of breast cancer.

Data from the Iowa Women's Health Study showed no increased risk of breast cancer in women who had used HT versus those who had not taken hormones(113). Additionally, when researchers went back and analyzed data from women who had developed breast cancer, they found that HT, in a very small number of women, was associated with cancer with a favorable prognosis(114). This finding is supported by other studies which have shown that women who use HT are less likely to have metastatic disease, and have a longer life expectancy than women who have not used HT(19). The findings of these studies suggest that rather than acting as a carcinogen, estrogen may act as a mitogen. However, one possible explanation for these findings is that women on HT are more likely to be seeing a doctor regularly and to undergo regular breast examinations and mammograms.

Data from the Nurses' Health Study showed a survival advantage for women taking estrogen at the time their breast cancer was diagnosed. The increased survival rate was associated with a lower frequency of late-stage disease and undoubtedly reflects earlier diagnosis in estrogen users(19). However, other evidence suggests that estrogen users develop better differentiated tumors and that surveillance or detection bias is not the only explanation for better survival(115, 116).

A number of recent studies have aroused concern over the effect of menopausal HT on breast tissue density. In women not on HT, breast density has been found to be an independent risk factor for breast cancer(117). Hormone therapy has been found to increase breast density, with the greatest increase in women on conjugated estrogen and progesterone(118).

Although an association between breast density and breast cancer has not been seen in women on HT, there has been some concern that mammograms may be less effective in women on HT with greater breast density. However, Rutter et al. showed that two weeks after discontinuing HT, women's breast density returned to normal(119). Therefore, until this issue is better understood, it may be advisable for women to discontinue HT for two weeks before a mammogram exam, especially in the case of prior problematic mammograms.

Evidence suggests, however, that estrogen plus progestin may have an impact on breast cancer. In July 2002, the estrogen plus progestin arm of the Women’s Health Initiative study was stopped due to a small increase in the incidence of breast cancer among women taking this combination. This risk amounted to approximately 8 more women per 10,000 being diagnosed with breast cancer compared to those on placebo(13). It is important to note, however, that the average age of women in this study was 63.2 years and does not reflect women on HT in normal clinical practice. In addition, 50% of the women in WHI were either current or former smokers, they had an average BMI of 28 (well-above normal), and 1/3 suffered from hypertension.

In the MWS (Million Women Study), the large British study, women on HT followed for 2.6 years were found to have an increased risk of breast cancer (RR 1.66) (26). Various hormone preparations were tested in this trial and similar risks were reported for all types, suggesting that risks are not confined to the standard CEE/MPA dose used in WHI. It is important to note though, that women taking estrogen only had a significantly lower increase in risk compared with women taking both an estrogen and progestogen. It is an important to recognize that this was an observational study only and hence has a larger potential area for error.

Although there is some evidence that combination therapy may increase risk of breast cancer above that of estrogen alone, neither a protective, nor a detrimental effect has been demonstrated convincingly, particularly for younger, healthier women. One study interviewed nearly 4000 women with and without breast cancer and found a significant correlation between use of continuous combined replacement therapy and breast cancer(120). However, the risks were higher in thin women than in heavier women which may confound the results. Also, it is possible that the use of cyclic therapy could provide the additional risk, and HT was generally given at higher doses that are rarely used today.

While there has been little consistency among the findings of the various studies on the effects of menopausal HT on breast cancer, one issue that is consistent in the literature is the observation that mortality from breast cancer is decreased among ET/HT users. A summary of the literature from 1990-2001 shows the RR of mortality consistently to be <1.0 with HT use [75-80]. One hypothesis to explain this observation is that HT may promote the development of slow-growing tumors or discourage the development of more aggressive tumors. Hulley et al, reported that tumors in women taking /HT were smaller, had a better histologic differentiation, an a lower cell-proliferation rate compared to nonusers(121). It has also been posited that better screening of these women leads to lower mortality rates.

The argument that menopausal HT should not be given to women who have a personal history of breast cancer may seem reasonable based on evidence that breast cancer is a hormone responsive tumor. However, while women with a first-degree relative (mother, sister, or daughter) who has or had premenopausal breast cancer are at increased risk by virtue of their family history alone, their risk of breast cancer is not thought to be increased further by HT use. Eighty percent of women who develop breast cancer do not have a family history. Sellers et al., examined HT use and breast cancer risk in women with a family history of breast cancer and found no statistically significant increase in risk in past or current users, regardless of duration of use (113). This is supported by the findings of Rebbeck et al., who studied women who were carriers of the BRCA1 gene mutation (122). Bilateral prophylactic oophorectomy was associated with a 47% reduction in breast cancer risk in this population. HT use did not negate the observed reduction in cancer risk. Interestingly, studies of breast cancer survivors showed that women using HT had a lower risk of recurrence compared to survivors not using HT (123, 124).

Breast cancer incidence is thought to increase after hormone use and since WHI there has been much interest on the role of the progestin in combination with estrogen in contrast to the use of estrogen alone(13, 14, 109). In general most studies that have shown a small increase have shown more of an effect with the combination (26) and nurse health and collaborative study). This has led to speculation as to the role of progestin, and to the minimization of progestin use despite the well-recognized and significant risk of endometrial cancer with the use of unopposed estrogen. Some recent studies suggest the progesterone and dydrogesterone may be safer than other progestins but no randomized studies examine this question(125). In general, some effect is seen with treatment duration and some studies show an effect although small. WHI reported an increase in breast cancer risk in the combined therapy arm in subjects who had used hormones prior to enrollment but only after 5 years (109). A later paper from the WHI study however suggested that the risk was higher in women who initiated therapy soon after menopause (within 3 to 5 years) (90). However, in this study, a much larger group of women who were recently menopausal had been on HT and the effect was more pronounced in the less rigorous observational arm. In general the effect takes several years to appear and is small. When hormones are discontinued the effect starts to decline within one year (96). All of this confusing and contradictory data suggests that the combined HT may be acting as a promoter in susceptible women with undiagnosed subclinical cancer and the promoter effect may disappear with discontinuation of therapy. This may also explain the overall drop in breast cancer seen with the Seer (Surveillance, Epidemiology and End Result) cancer registries database report. This report showed a drop in breast cancer rates after 2002 when women stopped hormone therapy after the WHI publications(126). This effect has not been seen universally and the trend was actually seen prior to the reports. In fact there has been a drop in many different cancer rates, possibly due to earlier detection and earlier treatment(127). Another item of interest is that the use of the less common lobular cancer of the breast (approximately 16 % vs. 70% for more common ductal cancers) is increased with hormone use (128). However this effect was not seen in WHI. Both combined hormone use and estrogen alone lead to denser breasts and more abnormal mammograms (111, 129). This effect is rapidly reversible and stopping hormones 10 to 30 days before a mammography may decrease abnormalities requiring follow up(130). One group of women who benefit from hormone therapy is the women with BRAC 1 and 2 mutations who undergo oophorectomy as prophylaxis. Use of HT does not appear to place them at risk for the genetically determined breast cancer and will improve quality of life(131). It will also prevent the effects of estrogen deprivation at a young age. The effects of stopping hormones are contradictory depending on the study. The Nurse’s Health Study reports that the risk is no longer present after 5 years while follow up in The WHI study shows a persistence of effect after 11 years of follow up(132).

Breast cancer prognosis does not appear to be influenced by the high hormone levels during pregnancy, nor has oral contraceptive use been shown to increase breast cancer risk. These observations may allay some of the fear regarding the use of exogenous hormones after menopause

OVARIAN CANCER

Data on ovarian cancer has not shown a consistent risk with use of hormone therapy. There is possible weak association with long term (at least 10 years) of therapy but data are inconclusive for recommendations (133). Its use does not adversely affect the risk of cancer in BRCA mutations (134). While WHI researchers reported an increased risk of ovarian cancer (HR 1.58), it did not reach statistical significance (135). Other studies too, including HERS and a meta-analysis of 15 case-controlled studies found no significant association(135-137).

ENDOMETRIAL CANCER

In 2001, 38,300 cases of endometrial cancer were diagnosed, and 6,600 women died of the disease. The mean age at diagnosis is 61 years, with most cases occurring in women 50 to 59 years old.

Estrogen alone causes endometrial hyperplasia and a two to three-fold increase in the risk of endometrial cancer. However, the addition of progestogen reduces this risk to lower levels than those seen in women not on HT(138, 139). Thus, the addition of a progestational agent to postmenopausal estrogen therapy is now standard for women with an intact uterus. While there have been some reports that the risk of endometrial cancer may be slightly increased even with the combined therapy, most studies have not confirmed this. Women in the WHI study on combined therapy showed no difference in endometrial cancer rates compared to women on placebo (13). Recent research has focused on the use of lower doses of estrogen and a progestogen in HT to reduce the risk of endometrial cancer(140).

The dose of progestogen given depends on several factors, including the number of days given each month, the amount of estrogen given, the individual needs of the patient, and her ability to tolerate the medication. Side effects of progestogen can include anxiety, irritability, depressed mood, acne, bloating, fluid retention, headaches, breast tenderness, and bleeding problems. The inability to tolerate these effects is the main reason for poor compliance or discontinuation of HT.

COLON CANCER

Despite being one of the major causes of cancer-related mortality in women, colon cancer is often overlooked by patients in their risk assessment of HT. Case-controlled and cohort studies have both found a 50% decrease in relative risk of colon cancer in women who are current or long-term HT users compared to women not on HT. In addition, reports from the WHI study showed that the combined estrogen plus progestin therapy was associated with a decrease in the incidence of colon cancer compared to women on placebo (6 fewer cases per 10,000 women on HT(13). This was not found with estrogen alone(14). Although the exact mechanism of estrogen and progestin’s protective effect on the colon is unclear, it has been suggested that estrogen acts to decrease bile acids, which are thought to be carcinogenic. At present; however, although the evidence that HT may be beneficial in reducing the risk of colon cancer should be considered, there is insufficient evidence to warrant recommending long-term HT solely for this purpose.

NEUROLOGIC FUNCTION

Cognition

The existence of estrogen receptors in the hippocampus, a part of the brain essential to learning and memory, has been known for some time. Several mechanisms may account for the effects of estrogen on the brain. Firstly, estrogen increases levels of choline O-acetyl-transferase, the enzyme needed to synthesize acetylcholine, a neurotransmitter thought to be critical for memory(141). Studies on healthy middle-aged and elderly postmenopausal women have supported the theory that estrogen may help to maintain aspects of cognitive function(142),(143). Data also suggest that estrogen therapy may enhance short- and long-term memory(144),(145). Additional effects of estrogen on neural function include: protecting neurons from oxidative stress and glutamate toxicity (146),(147), increasing glucose transport and cerebral blood flow , and stimulating the branching of neurites (148). A recent review of clinical trials of hormone therapy suggest that there is a clear difference between the effects of estrogen therapy and estrogen plus progestin (149). There is modest support for the beneficial effect of estrogen alone on verbal memory in women under 65, and possibly surgically menopausal, while a harmful effect is seen with estrogen plus progestin in women over 65. Conjugated estrogen with medroxyprogesterone acetate may also have some detrimental effect on younger women. Estrogen alone appears to be neutral in women over 65. Thus the age of initiation of therapy and the use of progestins are important when evaluating possible effects on verbal memory(149). At present there is no combination which appears to be neutral to verbal memory and there is suggestion of some harm even with micronized progesterone (150). Hot flashes appear to relate to memory dysfunction, and some of the cognitive improvement on hormone therapy may relate to the treatment of the hot flashes(151).

Alzheimer's Disease

For every five years after the age of 65, the prevalence of Alzheimer's disease doubles in the population. Nearly 50% of women over the age of 75 may suffer from the condition(152). As the population ages over the next 20 years, these numbers are expected to increase.

According to epidemiologic evidence, there is reason to believe that estrogen deficiency may contribute to Alzheimer's disease. Low body weight is associated with low levels of circulating estrogens in postmenopausal women. Women who suffer from Alzheimer's disease tend to have lower body weights than women without the disorder(153). Incidences of Alzheimer's disease are low or its expression is delayed in postmenopausal women with high levels of endogenous estrogenic steroids or those receiving long-term HT.

One explanation for estrogen's apparent protective effect may involve neurotransmission. Estrogen acts as a trophic factor for cholinergic neurons in vitro. Cholinergic depletion is the most prominent neurotransmitter deficit in Alzheimer's disease.

With regard to the association between risk of Alzheimer’s Disease and HT use, however, there is little consistency in the literature. However, while HT does show promise in preventing or delaying the onset of the disease, a recent study showed no benefit of either 0.625 mg/d or 1.25 mg/d of estrogen on Alzheimer's progression(154). Most likely, estrogen may merely delay the deterioration seen in Alzheimer's patients. Paganini-Hill and Henderson(155) reported a 35% decrease in risk for ET users compared to placebo, and Zandi et al.,(156) reported a 41% reduced risk for ever users of HT. However, the results from the WHIMS, the Women’s Initiative Memory Study, a substudy of WHI, reported that while HT did not significantly increase the risk of mild cognitive impairment (HR 1.07), it did increase the risk of probable dementia (HR 2.05) (157). The effect of HT on different subtypes of dementia could not be determined because the number of cases was too small. It must be noted, however, that because the WHIMS participants were all 65 or older, these results may not apply to women who initiate HT at a younger age.

The results of the Cache County Study(156) serve to further confuse the issue. In this prospective study of incident dementia in older women (mean age 74.5 years), the risk of AD was increased in current HT users with 10 or fewer years of therapy (HR 2.41 for fewer than 3 years of therapy, 2.12 for 3-10 years). For current users with more than 10 years of therapy the HR was 0.55, indicating a decrease in risk, but this value did not reach statistical significance. Interestingly, in past users, reductions were present in all age groups and showed a duration effect (HR 0.58 for fewer than 3 years, 0.32 for 3-10 years, and 0.17 for more than 10 years).

OTHER POSSIBLE RISKS

Thromboembolic disease

The Nurses' Health Study showed a twofold increase in the risk of pulmonary embolism among postmenopausal women who were current estrogen users. The recent findings of the WHI study confirmed these findings for women on combined estrogen plus progestin therapy. Women on this treatment suffered 8 more pulmonary emboli per 10,000 than women on placebo(13). Although estrogen use has been associated with an increase in the relative risk of venous thromboembolism (VTE), the absolute risk remains low, as VTE occurs infrequently in this setting. Women on combined estrogen-progestin therapy in the WHI study suffered 18 cases of more venous thromboembolism than women on placebo. However, when considered against a 50% reduction in cardiovascular disease risk, the increased risk of VTE does not contraindicate estrogen replacement. It does, however, show that patients should be screened for a history of idiopathic thrombosis as this has been a consistent finding (22).

Gallbladder disease

Some epidemiologic studies have found an increased risk of gallstones among women who use HT. Estrogen has been shown to increase cholesterol saturation of bile, alter bile acid composition, and decrease bile flow. Each of these effects can enhance gallstone formation. Data from the Nurses' Health Study (54,845 postmenopausal women monitored for eight years) showed that current HT users were more likely to have undergone cholecystectomy than nonusers (relative risk, 2.1). This risk tends to increase with long-term therapy and with high doses of estrogen(158).

Weight gain

Because many women gain weight as they age, a common fear is that HT will exacerbate this problem. However, this is unconfirmed by prospective studies. The PEPI trial showed that women on HT gained less weight than women not taking hormones(101). Attention to diet (with reduced fat intake) and regular aerobic exercise for weight maintenance should be recommended to all postmenopausal women. Data from WHI also showed an attenuation of increases in weight seen with age in the combined hormone treated arm(159). This suggests there may be some beneficial effect to HT on the normal increases that are seen in postmenopausal women and that the effect may protect against the increase in central obesity seen in hypoestrogenic menopausal women. A decrease in the incidence of diabetes, and lower insulin levels suggestive of better insulin sensitivity may be related to this attenuated weight gain.(160).

OTHER POSSIBLE BENEFITS

About 35% of patients over the age of 75 are affected by macular degeneration, the leading cause of severe vision loss in the elderly. One study showed that women who experienced menopause earlier in life had a 90% increased risk of developing symptoms of age-related macular degeneration later in life as compared to women who underwent menopause at an older age(161). Some studies have shown a small reduction in the incidence of this eye disorder among users of HT(162, 163).

Skin

It is thought that skin may be an important target organ for reproductive hormones. In postmenopausal women, dermal collagen decreases, and skin becomes thinner. Applying estrogen cream to the skin after menopause improves the external appearance of facial skin. In addition, systemic HT increases dermal collagen and limits age-related skin extensibility. To date, of the eleven clinical trials that examined the effect of HT on collagen levels, only one failed to demonstrate efficacy (164). Furthermore, results from a recent study indicates that estrogen also increases skin thickness (165).

HT has also been shown to accelerate cutaneous wound healing, both microscopically and macroscopically, in postmenopausal women (166). This study also showed delayed repair of acute incisional wounds in ovariectomized young female rodents; the delay was reversed by the topical application of estrogen.

Tooth loss

The risk of tooth loss increases after menopause. Osteoporosis, as well as estrogen deficiency, could both be contributing to this effect. Data from the Nurses' Health Study indicate that the risk of tooth loss may be decreased in women with a history of estrogen therapy (167).

TREATMENT

Non hormonal and combination treatments

Several treatments have recently become available and have FDA approval for relief of vasomotor symptoms. This includes a selective serotonin reuptake inhibitor, low dose paroxetine(168, 169) and basedoxifene/conjugated estrogens which also affords protection of bone. The latter consists of a combination of CEE and a SERM and is indicated for women with a uterus. A progestin is not necessary as this combination offers endometrial safety(170-172), Another SERM ospemifene has been approved for the treatment of postmenopausal vulvovaginal atrophy (173-175). Another treatment consists of a Swedish pollen extract femal, which has been shown to be effective in a small study for a composite of menopausal symptoms including vasomotor symptoms, fatigue and quality of life(176).

HORMONE THERAPY PRINCIPLES

Over the years, doses of estrogen in hormone therapy have been decreasing: until the mid-1970s, daily doses of CE as high as 1.25 or 2.5 mg were commonly used. Today, a CE dose of 0.625 mg/day is considered the “standard” dose for estrogen therapy while many women have relief of symptoms with even lower doses.

The goal of hormone therapy is to reduce menopausal symptoms (e.g., vasomotor symptoms, sleep disturbance, vulvovaginal symptoms, decreased libido) using the lowest effective dose for the shortest amount of time. Use of the lowest clinically effective dose of HT for relief of menopause-related symptoms and for prevention of osteoporosis is now recommended. Low-dose estrogen therapy (ET) is currently defined as a dose of oral CEE of ≤0.45 mg/d, oral estradiol ≤0.5mg/d, transdermal estradiol ≤0.0.375 mg/d, or the equivalent. The benefit-risk ratio of hormone therapy for each woman is influenced by the severity of her menopausal symptoms and their impact on quality of life, her current age, age at menopause, time since menopause, cause of menopause, and baseline disease risks. Some patients may require “standard” doses; however, and doses can be reduced if desired after 6 months to a year.

Generally appropriate indications include also treatment or prevention of osteoporosis in women who are not candidates for (or cannot tolerate) other osteoporosis therapies including bisphosphonates or teriparatide.

Absolute contraindications for systemic HT include hormone-related cancer, active liver disease, history of hormone-induced venous thromboembolism, history of pulmonary embolism not caused by trauma, vaginal bleeding of unknown etiology, and pregnancy. Relative contraindications include chronic liver disease, severe hypertriglyceridemia, endometriosis, history of endometrial cancer, history of breast cancer, coronary artery disease.

Guidelines for hormone use are reviewed in the statement of the North American Menopause Society(177) and recently by the Endocrine Society(178).

Considerable confusion has developed as a result of the numerous transdermal preparations which have appeared on the market. The effective dose depends on the delivery rate and the surface area applied so that there is much variation in terms of estradiol delivered to the blood stream. The following charts attempt to present equivalent doses. Lower doses take longer (4-7 weeks) for effective relief, and it is important to individualize therapy. Most preparations take a full 12 weeks for maximum effect although standard therapy provides relief sooner (2-3 weeks). There is also much debate as to the safety of oral vs. transdermal estrogen and the issue of dose vs delivery has not been resolved by double blind randomized trials. One study suggests that venous thromboembolism may be lower with transdermal products, but the doses compared were not equivalent(179). Another study shows a decreased risk of stroke in women on transdermal preparations with higher doses of both oral and transdermal estrogen showing significant effect (180). One study suggests progesterone may be associated with a lower risk of breast cancer than progestins but this again awaits further study (181).

Bioidentical Hormones.

The unfortunate publicity concerning compounded hormones mislabeled as ‘bioidentical” has suggested that custom made preparations based on saliva or blood levels were safer or better tolerated has lead to a cottage industry which has no scientific basis. These preparations offer no advantage over regulated and tested preparations approved by the FDA, and their risk is equivalent to commercial compounds. Claims that they are safer are misleading particularly since they have not been studied and one of the estrogens used, estriol, has no safety or efficacy data. Prescribers who claim they are more “natural “ do not inform patients that they are synthesized from plant chemicals extracted from yams or soy similar to some commercial preparations.

In general initiation of treatment of the symptomatic newly menopausal women will provide benefit which greatly outweighs risk and provides protection from bone loss. Older women who continue to be symptomatic may. continue treatment preferably with lower doses.

TREATMENT GUIDELINES

Although the decision to treat menopausal women rests on individualized risk vs. benefit for the patient some helpful clinical guidelines are useful for the clinician. In general, hormone treatment is being used for symptoms. These include vasomotor symptoms and vulvovaginal atrophy. There are, however, a variety of symptoms which make up the menopausal syndrome and are not strictly classified as vasomotor or vulvovaginal symptoms and are distressing to the patient and may also be a consideration for treatment(182). These include: mood disorders appearing at the perimenopause and menopause, migraines, severe insomnia, anxiety, difficulty concentrating, memory issues, severe fatigue and somatic symptoms especially joint pains and rarely muscle pain or a generalized crawling feeing on the skin. Some patients can endure two hot flashes a day while others who are in stressful or public jobs cannot. Patients are usually uncomfortable and distressed by more than two hot flashes per day. In particular the patient who wakes at night two or more times and suffers from sleep deprivation is usually in need of treatment. Patients suffering from five to seven hot flashes a day are experiencing moderate to severe symptoms and should be offered treatment. The physician should help the patient make a quality of life decision and advise these patients on the low risks associated with treatment particularly for a few years. Some patients may be experiencing bone loss and hormone therapy is ideal for this type of patient. Some of the estrogens on the market are also approved for prevention of osteoporosis and data shows they are very effective and prevent fractures. A patient on hormone therapy does not need a second drug for prevention of bone loss. If bone loss is occurring on hormone therapy a secondary cause should be searched for such as vitamin D deficiency, over treatment with thyroid hormone or hyperparathyroidism. Patients with mood issue may have problems with progestins and micronized progesterone or a vaginal delivery system may be better tolerated. Estrogens should be started first and a progestin added after a few weeks. Patients with migraines also have special tolerability issues and fluctuations of hormone levels which may be triggering the headaches may persist or be aggravated initially by hormone treatment. A transdermal patch may be the best option and a progestin should be started after a trial of treatment with estrogen. The issue of duration of hormone treatment will arise. Two to five years is usual. The small risk of breast cancer is also important to review with the patient. This risk surfaces after 5 years of use and did not surface at all with estrogen alone therapy after 7 seven years. This interval does not apply to patients with premature menopause who have been shown to be at risk for osteoporosis and premature heart disease if they are not replaced. All patients need a yearly mammogram and the increase in density can be avoided by stopping hormones for two weeks prior to the mammogram if she can tolerate it. Some patients stay on hormone therapy long term because of mood or other issues or they are in the unfortunate 10 percent who continue to suffer form vasomotor symptoms or cannot tolerate other drugs for osteoporosis. Patients with severe mood issues may require antidepressants. Recent data has shown the efficacy of low doses for vasomotor symptoms and many are available. However the patient with severe symptoms may prefer a standard dose which may be lowered after 6 months when symptoms are well controlled. Lastly, vaginal estrogens are an excellent option for patients with symptoms of vaginal atrophy and do not have the risks associated with systemic use. In particular, recurrent urinary tract infections and or vulvovaginitis are a hallmark of genitourinary estrogen deficiency which can be easily relieved or prevented with the use of vaginal estrogen. Treatment with hormone therapy is very individualized and quality of like may be greatly improved its use. When therapy is discontinued, a return of symptoms is common(183) although generally in a milder form. Unfortunately there is little data to guide the physician but many clinicians slowly taper doses over several months.

When assessing risk vs. benefit for long-term risks, the following conclusions from the WHI should be taken into consideration (13, 14, 88):

 

Over 1 year, per 10,000 women, estrogen/progestogen treated women had the following observed differences compared to controls:Estrogen and Progestin Estrogen Alone

  • 7 more CHD Events 5 fewer CHD events
  • 8 more strokes 12 more strokes
  • 8 more invasive breast cancers 7 fewer breast cancers
  • 18 more VTEs 7 more VTE
  • 8 more PEs 3 more PEs
  • 6 fewer colorectal ca 1 more colonrectal ca
  • 5 fewer hip fractures 6 fewer hip fractures
  • Long term follow up of subjects in WHI after discontinuation of treatment(132):
  • Intervention: CEE+MPA RR
  • Breast1.24, Stroke 1.37, Pulm embolism1.98
  • Colorectal CA 0.62, hip fracture 0.67, diabetes 0.81
  • Post Intervention :CEE + MPA.
  • Breast 1.28 all others attenuated
  • Intervention: CEE RR
  • Stroke 1.35, Hip fracture 0.67, diabetes 0.87
  • Post intervention: CEE RR
  • Breast CA 0.70, under 60 favorable mortality, less MI
  • Overall mortality not affected

 

 

 

1. Assess patient’s risk and symptoms.
Risks Symptoms
Osteoporosis
  • Amenorrhea or missed menstrual periods
  • Hot flashes or night sweats
Cardiovascular disease
  • Urogenital symptoms
  • Decreased sex drive, libido
Surgical menopause
  • Insomnia
  • Dyspareunia
Premature menopause
  • Osteoporotic-related height loss, disability, pain
  • Depression, mood change
Family history of Alzheimer’s disease
  • Headache
  • Irritability, emotional lability

 

 

 2. If risk or symptoms are present, screen for HT appropriateness.

CONTRAINDICATIONS

Absolute Relative
Hormone-related cancer or active liver disease Chronic liver disease
History of hormone-induced thromboembolism Severe hypertriglyceridemia
History of pulmonary embolism not caused by trauma Endometriosis
Vaginal bleeding of unknown etiology History of endometrial cancer
Pregnancy History of breast cancer
Proven coronary heart disease or recent event

 

 

3. If not appropriate, consider alternative therapies.

 

 

4. If appropriate for HT, consider the following:

ESTROGEN ALONE IF UTERUS IS ABSENT OR ESTROGEN-PROGESTOGEN IF UTERUS PRESENT

ESTROGEN/ANDROGEN (E/A) THERAPY

Risks present for: Symptoms present: Risks present for: Symptoms present:
Osteoporosis
  • Hot flashes or night sweats
Osteoporosis, not responsive to HT Symptoms as for ERT and/or:

  • Low energy
  • Decreased sex drive, libido
  • Muscle wasting
Cardiovascular disease
  • Urogenital symptoms
  • Osteoporotic-related height loss, disability, pain
Surgical menopause
  • Insomnia
Premature menopause
  • Headache
  • Irritability, emotional lability
  • Depression, mood change
  • Dyspareunia

 

 

5. After starting ET therapy, re-evaluate at 3 to 6 months.
  • If symptoms are controlled, continue HT.
  • If symptoms are not controlled or undesirable side effects are present:
Symptoms:
  • Headaches
  • Breast Pain
  • Urogenital symptoms
  • Irritability, emotional lability
  • Persistent hot flashes or night sweats
  • Decreased sex drive, libido
  • Fatigue
  • Insomnia
  • Depression, mood change
  • Irritability, emotional lability
  • Headache

 

Treatment:
  • Re-evaluate HT (dose and/or type)
  • Consider lower dose
  • Re-evaluate (3-6 months)
  • If symptoms controlled, continue treatment.
  • If symptoms not controlled or undesirable side effects persist, consider E/A or alternative therapy or consultation.
  • Issues of treatment duration will vary with individualized consideration of osteoporosis, dementia, or emerging issues. Breast cancer risk is present and is very small, less than 1/1000 women or less than 0.1%. Lower doses can be considered after symptoms are controlled. Time frame to consider lower doses should be individualized.

 

 6. Hormone Products for Treatment of Menopausal Symptoms
Estrogen Preparations
Doses (standard) Low dose Therapy
  • Conjugated (equine and synthetic) estrogens: 0.625, 0.9, 1.25 mg,
  • Micronized estradiol: 1, 2 mg
  • Ethinyl estradiol: 5 μg
  • Estradiol valerate: 2 mg
  • Piperzaine estrone sulfate: 0.625,1.25, 2.5 mg
  • Esterified estrogen: 0.625, 1.25, 2mg
  • estradiol acetate 0.9, 1.8 mg
  • Conjugated (equine and synthetic) estrogens: 0.3, 0.45 mg
  • Micronized estradiol: 0.5mg
  • Ethinyl estradiol: N/A
  • Estradiol valerate: N/A
  • Piperzaine estrone sulfate: N/A
  • Estradiol acetate 0.45 mg

 

Transdermal
  • E2 patch to deliver 0.05 mg/d
  • E2 patch to deliver 0.0375 ,0025,0.014 mg /day
  • E2 gel
  • to deliver 0.035, 0.025, ------------------
  • 0.027,0.0125,0.009,0.003/day
  • E spray
  • To deliver 0.021 mg/day

 

 

Vaginal Preparations
  • Conjugated Estrogen Cream
  • Estradiol Vaginal Cream
  • Estradiol Vaginal Tablets 25 or 10μg
  • Estrogen Vaginal Ring 2 mg
Treatment regimen:
  • Continuous
  • Cyclic

 

 

Progestin and Progesterone doses and types; The doses are for standard estrogen regimens. Doses can be halved with half doses and must be increased with higher estrogen doses. In general doses of estrogen therapy producing 35 to 60 pg/ml serum levels require standard doses of progestin but little literature is available.
CYCLIC
Medroxyprogesterone acetate: 5-10 mg for days 1-14 of each month
Norethindrone acetate: 2.5 mg for days 1-14 of each month
Micronized progesterone: 100 mg a.m. and 200 mg p.m. for days 1-14 of each month or 200mg in p.m., 2 hours after a meal
COMBINED (Use half with Half doses)
Medroxyprogesterone acetate: 2.5mg or 5mg daily
Micronized progesterone: 100 mg daily in the p.m., 2 hours after a meal
Other
Vaginal progesterone 4% : 6 doses every other day monthly. 45 mg per applicator (not FDA approved for menopausal use)
Levonorgestrel containing IUD 20ug/day release -5 year use (not FDA approved for menopausal use)

 

Combination EPT products
Cyclic
Regimen Route Available dose combinations
Conjugated Estrogen+medroxyprogesterone acetate :E alone for 14 days then E+P days 15-28 Oral, once a day 0.625 E+2.5 mg P0.625 E+5.0 mg P(2 tablets: E and E+P)
COMBINED
Regimen Route Available dose combinations
Conjugated equine estrogens (CEE) + medroxyprogesterone (MPA) Oral, once per day 0.625 mg CEE + 2.5 mg MPA, 0.625 mg CEE + 5.0 mg MPA, 0.45 or 0.3mg CEE +1.5mg MPA
Ethinyl estradiol (EE) + norethindrone acetate (NA) Oral, once per day 5 g EE + 1 mg NA, 2.5 ug EE + 1mg NA
17B-estradiol+norethindrone acetate 1MG e+0.5 MG na,0.5 mg E + 0.1MG NA,
17β-estradiol (E) +drospirenone (P) Oral, once a day 1 mg e=0.5 mg p
Oral intermittent combined
Micronized estradiol (E) + norgestimate (N) Oral, once/day 1 mg E + 0.09 mg NE (3 days on E/3 on E+P)

 

Transdermal Combinations
17β estradiol(E) + norethindrone acetate (P) One patch twice a week 0.05 mg E +0.25 mg P, 0.05 mg E +0.14 mg P,
17β estradiol (E)+ levonorgestrel (P) One patch once a week 0.45 mg E + 0.015 mg P
Micronized estradiol (ME) + norethindrone acetate (NA) Transdermal patch, replaced every 3-4 days 0.62 mg E + 2.7 mg NA
0.62 mg E + 4.8 mg NA

 

7. Therapeutic Hints:
  • Half doses of estrogen preparation can be used to decrease bleeding and breast tenderness. Recent data show good maintenance of bone with this approach.
  • Progestins may add to breast tenderness.
  • Moodiness and bloating may be due to progestins. Consider changing progestin, or using a lower dose of HT.
  • Consider using a patch in patients with high triglycerides to avoid “first pass” affect through the liver.
  • To improve HDL cholesterol use oral preparations. Increases are induced via a first pass mechanism.
  • Lower doses of HT/ERT have been found to be bone protective in doses equivalent to CEE 0.3mg.
  • Lower doses of HT can control vasomotor symptoms effectively when combined
    with progestin.
  • Lower doses or HT/ERT show favorable lipid profiles and changes are intermediate between standard dose and placebo.
  • Expect light bleeding or spotting in the first 3 months of therapy particularly with combined regimens. Abnormal bleeedoing (not the withdrawal bleeding after the progestin in a cyclic regimen should be evaluated with a pelvi/vaginal sonogram. If the endometrium is greater than 5mm an endometrial biopsy should be done to rule out hyperplasia.

 

REFERENCES

1. McKinlay SM, Brambilla DJ, Posner JG. The normal menopause transition. Maturitas. 1992;14(2):103-15.

2. Sevringhaus EL, Evans JS. Clinical observations on the use of an ovarian hormone: amniotin. Am J Med Sci. 1929;178:638-45.

3. Davis SR, Dinatale I, Rivera-Woll L, Davison S. Postmenopausal hormone therapy: from monkey glands to transdermal patches. J Endocrinol. 2005;185(2):207-22.

4. Ettinger B. Overview of estrogen replacement therapy: a historical perspective. Proc Soc Exp Biol Med. 1998;217(1):2-5.

5. Schmidt-Gollwitzer K. Estrogen/hormone replacement therapy present and past. Gynecol Endocrinol. 2001;15(suppl 4):11-6.

6. Nelson HD. Postmenopausal estrogen for treatment of hot flashes: clinical applications. JAMA. 2004;291(13):1621-5.

7. ASRM. The menopausal transition. Fertil Steril. 2008;90(5 Suppl):S61-5.

8. Longcope C, Franz C, Morello C, Baker R, Johnston CC, Jr. Steroid and gonadotropin levels in women during the peri-menopausal years. Maturitas. 1986;8(3):189-96.

9. Harlow SD, Gass M, Hall JE, Lobo R, Maki P, Rebar RW, et al. Executive summary of the Stages of Reproductive Aging Workshop + 10: addressing the unfinished agenda of staging reproductive aging. Menopause. 2012;19(4):387-95.

10. Gold EB, Bromberger J, Crawford S, Samuels S, Greendale GA, Harlow SD, et al. Factors associated with age at natural menopause in a multiethnic sample of midlife women. Am J Epidemiol. 2001;153(9):865-74.

11. Bachmann GA. Vasomotor flushes in menopausal women. Am J Obstet Gynecol. 1999;180(3 Pt 2):S312-6.

12. Haney AF, Wild RA. Options for hormone therapy in women who have had a hysterectomy. Menopause. 2007;14(3 Pt 2):592-7; quiz 8-9.

13. Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ, Kooperberg C, Stefanick ML, et al. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women's Health Initiative randomized controlled trial. JAMA. 2002;288(3):321-33.

14. Anderson GL, Limacher M, Assaf AR, Bassford T, Beresford SA, Black H, et al. Effects of conjugated equine estrogen in postmenopausal women with hysterectomy: the Women's Health Initiative randomized controlled trial. Jama. 2004;291(14):1701-12.

15. Manson JE, Hsia J, Johnson KC, Rossouw JE, Assaf AR, Lasser NL, et al. Estrogen plus progestin and the risk of coronary heart disease. N Engl J Med. 2003;349(6):523-34.

16. de Villiers TJ, Gass ML, Haines CJ, Hall JE, Lobo RA, Pierroz DD, et al. Global consensus statement on menopausal hormone therapy. Climacteric. 2013;16(2):203-4.

17. PEPI. Effects of estrogen or estrogen/progestin regimens on heart disease risk factors in postmenopausal women. The Postmenopausal Estrogen/Progestin Interventions (PEPI) Trial. The Writing Group for the PEPI Trial. JAMA. 1995;273(3):199-208.

18. Hulley S, Grady D, Bush T, Furberg C, Herrington D, Riggs B, et al. Randomized trial of estrogen plus progestin for secondary prevention of coronary heart disease in postmenopausal women. Heart and Estrogen/progestin Replacement Study (HERS) Research Group. JAMA. 1998;280(7):605-13.

19. Colditz GA, Egan KM, Stampfer MJ. Hormone replacement therapy and risk of breast cancer: results from epidemiologic studies. Am J Obstet Gynecol. 1993;168(5):1473-80.

20. Grodstein F, Stampfer MJ, Colditz GA, Willett WC, Manson JE, Joffe M, et al. Postmenopausal hormone therapy and mortality. N Engl J Med. 1997;336(25):1769-75.

21. Grodstein F, Manson JE, Stampfer MJ. Postmenopausal hormone use and secondary prevention of coronary events in the nurses' health study. a prospective, observational study. Ann Intern Med. 2001;135(1):1-8.

22. Grodstein F, Stampfer MJ, Goldhaber SZ, Manson JE, Colditz GA, Speizer FE, et al. Prospective study of exogenous hormones and risk of pulmonary embolism in women. Lancet. 1996;348(9033):983-7.

23. Finkelstein JS, Brockwell SE, Mehta V, Greendale GA, Sowers MR, Ettinger B, et al. Bone mineral density changes during the menopause transition in a multiethnic cohort of women. J Clin Endocrinol Metab. 2008;93(3):861-8.

24. Hess R, Colvin A, Avis NE, Bromberger JT, Schocken M, Johnston JM, et al. The impact of hormone therapy on health-related quality of life: longitudinal results from the Study of Women's Health Across the Nation. Menopause. 2008;15(3):422-8.

25. Avis NE, Crawford SL, Greendale G, Bromberger JT, Everson-Rose SA, Gold EB, et al. Duration of Menopausal Vasomotor Symptoms Over the Menopause Transition. JAMA Intern Med. 2015.

26. Beral V. Breast cancer and hormone-replacement therapy in the Million Women Study. Lancet. 2003;362(9382):419-27.

27. Vickers MR, MacLennan AH, Lawton B, Ford D, Martin J, Meredith SK, et al. Main morbidities recorded in the women's international study of long duration oestrogen after menopause (WISDOM): a randomised controlled trial of hormone replacement therapy in postmenopausal women. BMJ. 2007;335(7613):239.

28. Hodis HN, Mack WJ, Shoupe D, Azen SP, Stanczyk FZ, Hwang-Levine J, et al. Methods and baseline cardiovascular data from the Early versus Late Intervention Trial with Estradiol testing the menopausal hormone timing hypothesis. Menopause. 2014.

29. Cohen LS, Soares CN, Vitonis AF, Otto MW, Harlow BL. Risk for new onset of depression during the menopausal transition: the Harvard study of moods and cycles. Arch Gen Psychiatry. 2006;63(4):385-90.

30. Schierbeck LL, Rejnmark L, Tofteng CL, Stilgren L, Eiken P, Mosekilde L, et al. Effect of hormone replacement therapy on cardiovascular events in recently postmenopausal women: randomised trial. BMJ. 2012;345:e6409.

31. Freeman EW, Sherif K. Prevalence of hot flushes and night sweats around the world: a systematic review. Climacteric. 2007;10(3):197-214.

32. Grady D. Clinical practice. Management of menopausal symptoms. N Engl J Med. 2006;355(22):2338-47.

33. Kronenberg F. Hot flashes: epidemiology and physiology. Ann N Y Acad Sci. 1990;592:52-86; discussion 123-33.

34. Nelson HD. Commonly used types of postmenopausal estrogen for treatment of hot flashes: scientific review. JAMA. 2004;291(13):1610-20.

35. Utian WH, Shoupe D, Bachmann G, Pinkerton JV, Pickar JH. Relief of vasomotor symptoms and vaginal atrophy with lower doses of conjugated equine estrogens and medroxyprogesterone acetate. Fertil Steril. 2001;75(6):1065-79.

36. Good WR, John VA, Ramirez M, Higgins JE. Comparison of Alora estradiol matrix transdermal delivery system with oral conjugated equine estrogen therapy in relieving menopausal symptoms. Alora Study Group. Climacteric. 1999;2(1):29-36.

37. Utian WH. Psychosocial and socioeconomic burden of vasomotor symptoms in menopause: a comprehensive review. Health Qual Life Outcomes. 2005;3:47.

38. Woods NF, Mitchell ES. Symptoms during the perimenopause: prevalence, severity, trajectory, and significance in women's lives. Am J Med. 2005;118 Suppl 12B:14-24.

39. Freedman RR, Roehrs TA. Effects of REM sleep and ambient temperature on hot flash-induced sleep disturbance. Menopause. 2006;13(4):576-83.

40. Freedman RR, Roehrs TA. Sleep disturbance in menopause. Menopause. 2007;14(5):826-9.

41. Schiff I, Regestein Q, Tulchinsky D, Ryan KJ. Effects of estrogens on sleep and psychological state of hypogonadal women. JAMA. 1979;242(22):2405-4.

42. Harlow BL, Wise LA, Otto MW, Soares CN, Cohen LS. Depression and its influence on reproductive endocrine and menstrual cycle markers associated with perimenopause: the Harvard Study of Moods and Cycles. Arch Gen Psychiatry. 2003;60(1):29-36.

43. Bromberger JT, Meyer PM, Kravitz HM, Sommer B, Cordal A, Powell L, et al. Psychologic distress and natural menopause: a multiethnic community study. Am J Public Health. 2001;91(9):1435-42.

44. Bromberger JT, Matthews KA, Schott LL, Brockwell S, Avis NE, Kravitz HM, et al. Depressive symptoms during the menopausal transition: the Study of Women's Health Across the Nation (SWAN). J Affect Disord. 2007;103(1-3):267-72.

45. Freeman EW, Sammel MD, Liu L, Gracia CR, Nelson DB, Hollander L. Hormones and menopausal status as predictors of depression in women in transition to menopause. Arch Gen Psychiatry. 2004;61(1):62-70.

46. Bromberger JT, Kravitz HM, Matthews K, Youk A, Brown C, Feng W. Predictors of first lifetime episodes of major depression in midlife women. Psychol Med. 2009;39(1):55-64.

47. Avis NE, Brambilla D, McKinlay SM, Vass K. A longitudinal analysis of the association between menopause and depression. Results from the Massachusetts Women's Health Study. Ann Epidemiol. 1994;4(3):214-20.

48. Dennerstein L, Guthrie JR, Clark M, Lehert P, Henderson VW. A population-based study of depressed mood in middle-aged, Australian-born women. Menopause. 2004;11(5):563-8.

49. Schmidt PJ, Haq N, Rubinow DR. A longitudinal evaluation of the relationship between reproductive status and mood in perimenopausal women. Am J Psychiatry. 2004;161(12):2238-44.

50. Schmidt PJ, Rubinow DR. Sex hormones and mood in the perimenopause. Ann N Y Acad Sci. 2009;1179:70-85.

51. Callegari C, Buttarelli M, Cromi A, Diurni M, Salvaggio F, Bolis PF. Female psychopathologic profile during menopausal transition: a preliminary study. Maturitas. 2007;56(4):447-51.

52. Steiner M, Dunn E, Born L. Hormones and mood: from menarche to menopause and beyond. J Affect Disord. 2003;74(1):67-83.

53. Joffe H, Hall JE, Soares CN, Hennen J, Reilly CJ, Carlson K, et al. Vasomotor symptoms are associated with depression in perimenopausal women seeking primary care. Menopause. 2002;9(6):392-8.

54. Woods NF, Smith-DiJulio K, Percival DB, Tao EY, Mariella A, Mitchell S. Depressed mood during the menopausal transition and early postmenopause: observations from the Seattle Midlife Women's Health Study. Menopause. 2008;15(2):223-32.

55. Steinberg EM, Rubinow DR, Bartko JJ, Fortinsky PM, Haq N, Thompson K, et al. A cross-sectional evaluation of perimenopausal depression. J Clin Psychiatry. 2008;69(6):973-80.

56. Harlow BL, Signorello LB. Factors associated with early menopause. Maturitas. 2000;35(1):3-9.

57. Gyllstrom ME, Schreiner PJ, Harlow BL. Perimenopause and depression: strength of association, causal mechanisms and treatment recommendations. Best Pract Res Clin Obstet Gynaecol. 2007;21(2):275-92.

58. Soares CN, Almeida OP, Joffe H, Cohen LS. Efficacy of estradiol for the treatment of depressive disorders in perimenopausal women: a double-blind, randomized, placebo-controlled trial. Arch Gen Psychiatry. 2001;58(6):529-34.

59. Morrison MF, Kallan MJ, Ten Have T, Katz I, Tweedy K, Battistini M. Lack of efficacy of estradiol for depression in postmenopausal women: a randomized, controlled trial. Biol Psychiatry. 2004;55(4):406-12.

60. Harman M. Primary findings of the Kronos Early Prevention Study (KEEPS). Proceedings from the 23rd Anuual Meeting of the North American Menopause Society. Orlando Florida; 2012.

61. Parry BL. Perimenopausal depression. Am J Psychiatry. 2008;165(1):23-7.

62. Panay N, Studd J. Progestogen intolerance and compliance with hormone replacement therapy in menopausal women. Hum Reprod Update. 1997;3(2):159-71.

63. Sherwin BB, Gelfand MM. Sex steroids and affect in the surgical menopause: a double-blind, cross-over study. Psychoneuroendocrinology. 1985;10(3):325-35.

64. Sherwin BB. Affective changes with estrogen and androgen replacement therapy in surgically menopausal women. J Affect Disord. 1988;14(2):177-87.

65. Barlow DH, Samsioe G, van Geelen JM. A study of European womens' experience of the problems of urogenital ageing and its management. Maturitas. 1997;27(3):239-47.

66. Oriba HA, Maibach HI. Vulvar transepidermal water loss (TEWL) decay curves. Effect of occlusion, delipidation, and age. Acta Derm Venereol. 1989;69(6):461-5.

67. Pinkerton JV, Stovall DW, Kightlinger RS. Advances in the treatment of menopausal symptoms. Womens Health (Lond Engl). 2009;5(4):361-84; quiz 83-4.

68. Raymundo N, Yu-cheng B, Zi-yan H, Lai CH, Leung K, Subramaniam R, et al. Treatment of atrophic vaginitis with topical conjugated equine estrogens in postmenopausal Asian women. Climacteric. 2004;7(3):312-8.

69. Bachmann G, Lobo RA, Gut R, Nachtigall L, Notelovitz M. Efficacy of low-dose estradiol vaginal tablets in the treatment of atrophic vaginitis: a randomized controlled trial. Obstet Gynecol. 2008;111(1):67-76.

70. Biglia N, Peano E, Sgandurra P, Moggio G, Panuccio E, Migliardi M, et al. Low-dose vaginal estrogens or vaginal moisturizer in breast cancer survivors with urogenital atrophy: a preliminary study. Gynecol Endocrinol.

71. Santen RJ, Pinkerton JV, Conaway M, Ropka M, Wisniewski L, Demers L, et al. Treatment of urogenital atrophy with low-dose estradiol: preliminary results. Menopause. 2002;9(3):179-87.

72. Management of symptomatic vulvovaginal atrophy: 2013 position statement of The North American Menopause Society. Menopause. 2013;20(9):888-902; quiz 3-4.

73. Sherwin BB, Gelfand MM. The role of androgen in the maintenance of sexual functioning in oophorectomized women. Psychosom Med. 1987;49(4):397-409.

74. Cummings SR, Black DM, Rubin SM. Lifetime risks of hip, Colles', or vertebral fracture and coronary heart disease among white postmenopausal women. Arch Intern Med. 1989;149(11):2445-8.

75. Soyka LA, Fairfield WP, Klibanski A. Clinical review 117: Hormonal determinants and disorders of peak bone mass in children. J Clin Endocrinol Metab. 2000;85(11):3951-63.

76. Riggs BL, Wahner HW, Melton LJ, 3rd, Richelson LS, Judd HL, Offord KP. Rates of bone loss in the appendicular and axial skeletons of women. Evidence of substantial vertebral bone loss before menopause. J Clin Invest. 1986;77(5):1487-91.

77. Recker R, Lappe J, Davies K, Heaney R. Characterization of perimenopausal bone loss: a prospective study. J Bone Miner Res. 2000;15(10):1965-73.

78. FDA. Questions and answers for estrogen and estrogen with progestin therapies for postmenopausal women (updated) http://www.fda.gov/Drugs/DrugSafety/InformationbyDrugClass/ucm135339.htm accessed 3/29/2010. 2010.

79. Corson SL. A practical guide to prescribing estrogen replacement therapy. Int J Fertil Menopausal Stud. 1995;40(5):229-47.

80. Manson JE, Bassuk SS. Calcium supplements: do they help or harm? Menopause. 2014;21(1):106-8.

81. Ross AC, Manson JE, Abrams SA, Aloia JF, Brannon PM, Clinton SK, et al. The 2011 report on dietary reference intakes for calcium and vitamin D from the Institute of Medicine: what clinicians need to know. J Clin Endocrinol Metab. 2011;96(1):53-8.

82. Holick MF, Siris ES, Binkley N, Beard MK, Khan A, Katzer JT, et al. Prevalence of Vitamin D inadequacy among postmenopausal North American women receiving osteoporosis therapy. J Clin Endocrinol Metab. 2005;90(6):3215-24.

83. Anderson RN. Deaths: leading causes for 1999. Natl Vital Stat Rep. 2001;49(11):1-87.

84. Matthews KA, Meilahn E, Kuller LH, Kelsey SF, Caggiula AW, Wing RR. Menopause and risk factors for coronary heart disease. N Engl J Med. 1989;321(10):641-6.

85. Heckbert SR, Kaplan RC, Weiss NS, Psaty BM, Lin D, Furberg CD, et al. Risk of recurrent coronary events in relation to use and recent initiation of postmenopausal hormone therapy. Arch Intern Med. 2001;161(14):1709-13.

86. Alexander KP, Newby LK, Hellkamp AS, Harrington RA, Peterson ED, Kopecky S, et al. Initiation of hormone replacement therapy after acute myocardial infarction is associated with more cardiac events during follow-up. J Am Coll Cardiol. 2001;38(1):1-7.

87. Hodis HN, Mack WJ, Lobo RA, Shoupe D, Sevanian A, Mahrer PR, et al. Estrogen in the prevention of atherosclerosis. A randomized, double-blind, placebo-controlled trial. Ann Intern Med. 2001;135(11):939-53.

88. Rossouw JE, Prentice RL, Manson JE, Wu L, Barad D, Barnabei VM, et al. Postmenopausal hormone therapy and risk of cardiovascular disease by age and years since menopause. JAMA. 2007;297(13):1465-77.

89. Manson JE, Allison MA, Rossouw JE, Carr JJ, Langer RD, Hsia J, et al. Estrogen therapy and coronary-artery calcification. N Engl J Med. 2007;356(25):2591-602.

90. Prentice RL, Manson JE, Langer RD, Anderson GL, Pettinger M, Jackson RD, et al. Benefits and risks of postmenopausal hormone therapy when it is initiated soon after menopause. Am J Epidemiol. 2009;170(1):12-23.

91. Banks E, Canfell K. Invited Commentary: Hormone therapy risks and benefits--The Women's Health Initiative findings and the postmenopausal estrogen timing hypothesis. Am J Epidemiol. 2009;170(1):24-8.

92. Toh S, Hernandez-Diaz S, Logan R, Rossouw JE, Hernan MA. Coronary heart disease in postmenopausal recipients of estrogen plus progestin therapy: does the increased risk ever disappear? A randomized trial. Ann Intern Med;152(4):211-7.

93. Stevenson JC, Hodis HN, Pickar JH, Lobo RA. Coronary heart disease and menopause management: the swinging pendulum of HRT. Atherosclerosis. 2009;207(2):336-40.

94. Harman M. Primary findings of the Kronos Early Prevention Study (KEEPS). Proceedings from the 23rd Meeting of the North American Menopause Society. October 3-6 2012.

95. Hodis HN. Latest Data from the Elite Trial. International Menopause Society 14 World Congress on Menopause 2014. Cancun Mexico.

96. Heiss G, Wallace R, Anderson GL, Aragaki A, Beresford SA, Brzyski R, et al. Health risks and benefits 3 years after stopping randomized treatment with estrogen and progestin. JAMA. 2008;299(9):1036-45.

97. Bechlioulis A, Kalantaridou SN, Naka KK, Chatzikyriakidou A, Calis KA, Makrigiannakis A, et al. Endothelial function, but not carotid intima-media thickness, is affected early in menopause and is associated with severity of hot flushes. J Clin Endocrinol Metab;95(3):1199-206.

98. Matthews KA, Crawford SL, Chae CU, Everson-Rose SA, Sowers MF, Sternfeld B, et al. Are changes in cardiovascular disease risk factors in midlife women due to chronological aging or to the menopausal transition? J Am Coll Cardiol. 2009;54(25):2366-73.

99. Herrington DM, Reboussin DM, Brosnihan KB, Sharp PC, Shumaker SA, Snyder TE, et al. Effects of estrogen replacement on the progression of coronary-artery atherosclerosis. N Engl J Med. 2000;343(8):522-9.

100. Fahraeus L. The effects of estradiol on blood lipids and lipoproteins in postmenopausal women. Obstet Gynecol. 1988;72(5 Suppl):18S-22S.

101. Effects of estrogen or estrogen/progestin regimens on heart disease risk factors in postmenopausal women. The Postmenopausal Estrogen/Progestin Interventions (PEPI) Trial. The Writing Group for the PEPI Trial. JAMA. 1995;273(3):199-208.

102. Sarrel PM, Lindsay D, Rosano GM, Poole-Wilson PA. Angina and normal coronary arteries in women: gynecologic findings. Am J Obstet Gynecol. 1992;167(2):467-71.

103. Rosano GM, Sarrel PM, Poole-Wilson PA, Collins P. Beneficial effect of oestrogen on exercise-induced myocardial ischaemia in women with coronary artery disease. Lancet. 1993;342(8864):133-6.

104. Gangar KF, Vyas S, Whitehead M, Crook D, Meire H, Campbell S. Pulsatility index in internal carotid artery in relation to transdermal oestradiol and time since menopause. Lancet. 1991;338(8771):839-42.

105. Viscoli CM, Brass LM, Kernan WN, Sarrel PM, Suissa S, Horwitz RI. A clinical trial of estrogen-replacement therapy after ischemic stroke. N Engl J Med. 2001;345(17):1243-9.

106. Warren MP, Richardson, O, Chaundry,S.et al. The effect of Estrogen and Hormone Withdrawal on Health and Quality of Life after Publication of the Women's Health Initiative in New York City. Abstracts,Meeting of the North American Menopause Society Washington DC Sept. 2011;S-2:32.

107. Feuer EJ, Wun LM, Boring CC, Flanders WD, Timmel MJ, Tong T. The lifetime risk of developing breast cancer. J Natl Cancer Inst. 1993;85(11):892-7.

108. Anderson GL, Chlebowski RT, Aragaki AK, Kuller LH, Manson JE, Gass M, et al. Conjugated equine oestrogen and breast cancer incidence and mortality in postmenopausal women with hysterectomy: extended follow-up of the Women's Health Initiative randomised placebo-controlled trial. Lancet Oncol. 2012;13(5):476-86.

109. Anderson GL, Chlebowski RT, Rossouw JE, Rodabough RJ, McTiernan A, Margolis KL, et al. Prior hormone therapy and breast cancer risk in the Women's Health Initiative randomized trial of estrogen plus progestin. Maturitas. 2006;55(2):103-15.

110. Santen RJ, Song Y, Yue W, Wang JP, Heitjan DF. Effects of menopausal hormonal therapy on occult breast tumors. J Steroid Biochem Mol Biol. 2013;137:150-6.

111. Chlebowski RT, Hendrix SL, Langer RD, Stefanick ML, Gass M, Lane D, et al. Influence of estrogen plus progestin on breast cancer and mammography in healthy postmenopausal women: the Women's Health Initiative Randomized Trial. Jama. 2003;289(24):3243-53.

112. Breast cancer and hormone replacement therapy: collaborative reanalysis of data from 51 epidemiological studies of 52,705 women with breast cancer and 108,411 women without breast cancer. Collaborative Group on Hormonal Factors in Breast Cancer. Lancet. 1997;350(9084):1047-59.

113. Sellers TA, Mink PJ, Cerhan JR, Zheng W, Anderson KE, Kushi LH, et al. The role of hormone replacement therapy in the risk for breast cancer and total mortality in women with a family history of breast cancer. Ann Intern Med. 1997;127(11):973-80.

114. Gapstur SM, Morrow M, Sellers TA. Hormone replacement therapy and risk of breast cancer with a favorable histology: results of the Iowa Women's Health Study. JAMA. 1999;281(22):2091-7.

115. Bonnier P, Romain S, Giacalone PL, Laffargue F, Martin PM, Piana L. Clinical and biologic prognostic factors in breast cancer diagnosed during postmenopausal hormone replacement therapy. Obstet Gynecol. 1995;85(1):11-7.

116. Bergkvist L, Adami HO, Persson I, Bergstrom R, Krusemo UB. Prognosis after breast cancer diagnosis in women exposed to estrogen and estrogen-progestogen replacement therapy. Am J Epidemiol. 1989;130(2):221-8.

117. Boyd NF, Lockwood GA, Martin LJ, Knight JA, Byng JW, Yaffe MJ, et al. Mammographic densities and breast cancer risk. Breast Dis. 1998;10(3-4):113-26.

118. Greendale GA, Reboussin BA, Sie A, Singh HR, Olson LK, Gatewood O, et al. Effects of estrogen and estrogen-progestin on mammographic parenchymal density. Postmenopausal Estrogen/Progestin Interventions (PEPI) Investigators. Ann Intern Med. 1999;130(4 Pt 1):262-9.

119. Rutter CM, Mandelson MT, Laya MB, Seger DJ, Taplin S. Changes in breast density associated with initiation, discontinuation, and continuing use of hormone replacement therapy. JAMA. 2001;285(2):171-6.

120. Ross RK, Paganini-Hill A, Wan PC, Pike MC. Effect of hormone replacement therapy on breast cancer risk: estrogen versus estrogen plus progestin. J Natl Cancer Inst. 2000;92(4):328-32.

121. Holli K, Isola J, Cuzick J. Low biologic aggressiveness in breast cancer in women using hormone replacement therapy. J Clin Oncol. 1998;16(9):3115-20.

122. Rebbeck TR, Levin AM, Eisen A, Snyder C, Watson P, Cannon-Albright L, et al. Breast cancer risk after bilateral prophylactic oophorectomy in BRCA1 mutation carriers. J Natl Cancer Inst. 1999;91(17):1475-9.

123. Durna EM, Wren BG, Heller GZ, Leader LR, Sjoblom P, Eden JA. Hormone replacement therapy after a diagnosis of breast cancer: cancer recurrence and mortality. Med J Aust. 2002;177(7):347-51.

124. O'Meara ES, Rossing MA, Daling JR, Elmore JG, Barlow WE, Weiss NS. Hormone replacement therapy after a diagnosis of breast cancer in relation to recurrence and mortality. J Natl Cancer Inst. 2001;93(10):754-62.

125. Fournier A, Mesrine S, Boutron-Ruault MC, Clavel-Chapelon F. Estrogen-progestagen menopausal hormone therapy and breast cancer: does delay from menopause onset to treatment initiation influence risks? J Clin Oncol. 2009;27(31):5138-43.

126. Ravdin PM, Cronin KA, Howlader N, Berg CD, Chlebowski RT, Feuer EJ, et al. The decrease in breast-cancer incidence in 2003 in the United States. N Engl J Med. 2007;356(16):1670-4.

127. Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ. Cancer statistics, 2009. CA Cancer J Clin. 2009;59(4):225-49.

128. Li CI, Malone KE, Porter PL, Lawton TJ, Voigt LF, Cushing-Haugen KL, et al. Relationship between menopausal hormone therapy and risk of ductal, lobular, and ductal-lobular breast carcinomas. Cancer Epidemiol Biomarkers Prev. 2008;17(1):43-50.

129. Stefanick ML, Anderson GL, Margolis KL, Hendrix SL, Rodabough RJ, Paskett ED, et al. Effects of conjugated equine estrogens on breast cancer and mammography screening in postmenopausal women with hysterectomy. JAMA. 2006;295(14):1647-57.

130. Harvey JA, Pinkerton JV, Herman CR. Short-term cessation of hormone replacement therapy and improvement of mammographic specificity. J Natl Cancer Inst. 1997;89(21):1623-5.

131. Biglia N, Mariani L, Ponzone R, Sismondi P. Oral contraceptives, salpingo-oophorectomy and hormone replacement therapy in BRCA1-2 mutation carriers. Maturitas. 2008;60(2):71-7.

132. Manson JE, Chlebowski RT, Stefanick ML, Aragaki AK, Rossouw JE, Prentice RL, et al. Menopausal hormone therapy and health outcomes during the intervention and extended poststopping phases of the Women's Health Initiative randomized trials. Jama. 2013;310(13):1353-68.

133. Hormone therapy for the prevention of chronic conditions in postmenopausal women: recommendations from the U.S. Preventive Services Task Force. Ann Intern Med. 2005;142(10):855-60.

134. Kotsopoulos J, Lubinski J, Neuhausen SL, Lynch HT, Rosen B, Ainsworth P, et al. Hormone replacement therapy and the risk of ovarian cancer in BRCA1 and BRCA2 mutation carriers. Gynecol Oncol. 2006;100(1):83-8.

135. Anderson GL, Judd HL, Kaunitz AM, Barad DH, Beresford SA, Pettinger M, et al. Effects of estrogen plus progestin on gynecologic cancers and associated diagnostic procedures: the Women's Health Initiative randomized trial. JAMA. 2003;290(13):1739-48.

136. Hulley S, Furberg C, Barrett-Connor E, Cauley J, Grady D, Haskell W, et al. Noncardiovascular disease outcomes during 6.8 years of hormone therapy: Heart and Estrogen/progestin Replacement Study follow-up (HERS II). JAMA. 2002;288(1):58-66.

137. Coughlin SS, Giustozzi A, Smith SJ, Lee NC. A meta-analysis of estrogen replacement therapy and risk of epithelial ovarian cancer. J Clin Epidemiol. 2000;53(4):367-75.

138. Persson I, Yuen J, Bergkvist L, Schairer C. Cancer incidence and mortality in women receiving estrogen and estrogen-progestin replacement therapy--long-term follow-up of a Swedish cohort. Int J Cancer. 1996;67(3):327-32.

139. Gambrell RD, Jr. The menopause: benefits and risks of estrogen-progestogen replacement therapy. Fertil Steril. 1982;37(4):457-74.

140. Pickar JH, Yeh IT, Wheeler JE, Cunnane MF, Speroff L. Endometrial effects of lower doses of conjugated equine estrogens and medroxyprogesterone acetate: two-year substudy results. Fertil Steril. 2003;80(5):1234-40.

141. Bartus RT, Dean RL, 3rd, Beer B, Lippa AS. The cholinergic hypothesis of geriatric memory dysfunction. Science. 1982;217(4558):408-14.

142. Ditkoff EC, Crary WG, Cristo M, Lobo RA. Estrogen improves psychological function in asymptomatic postmenopausal women. Obstet Gynecol. 1991;78(6):991-5.

143. Kampen DL, Sherwin BB. Estrogen use and verbal memory in healthy postmenopausal women. Obstet Gynecol. 1994;83(6):979-83.

144. Sherwin BB. Estrogen effects on cognition in menopausal women. Neurology. 1997;48(5 Suppl 7):S21-6.

145. Resnick SM, Metter EJ, Zonderman AB. Estrogen replacement therapy and longitudinal decline in visual memory. A possible protective effect? Neurology. 1997;49(6):1491-7.

146. Behl C, Skutella T, Lezoualc'h F, Post A, Widmann M, Newton CJ, et al. Neuroprotection against oxidative stress by estrogens: structure-activity relationship. Mol Pharmacol. 1997;51(4):535-41.

147. Singer CA, Figueroa-Masot XA, Batchelor RH, Dorsa DM. The mitogen-activated protein kinase pathway mediates estrogen neuroprotection after glutamate toxicity in primary cortical neurons. J Neurosci. 1999;19(7):2455-63.

148. Dubal DB, Wilson ME, Wise PM. Estradiol: a protective and trophic factor in the brain. J Alzheimers Dis. 1999;1(4-5):265-74.

149. Maki PM, Sundermann E. Hormone therapy and cognitive function. Hum Reprod Update. 2009;15(6):667-81.

150. Pefanco MA, Kenny AM, Kaplan RF, Kuchel G, Walsh S, Kleppinger A, et al. The effect of 3-year treatment with 0.25 mg/day of micronized 17beta-estradiol on cognitive function in older postmenopausal women. J Am Geriatr Soc. 2007;55(3):426-31.

151. Maki PM, Drogos LL, Rubin LH, Banuvar S, Shulman LP, Geller SE. Objective hot flashes are negatively related to verbal memory performance in midlife women. Menopause. 2008;15(5):848-56.

152. Evans DA, Funkenstein HH, Albert MS, Scherr PA, Cook NR, Chown MJ, et al. Prevalence of Alzheimer's disease in a community population of older persons. Higher than previously reported. JAMA. 1989;262(18):2551-6.

153. Berlinger WG, Potter JF. Low Body Mass Index in demented outpatients. J Am Geriatr Soc. 1991;39(10):973-8.

154. Mulnard RA, Cotman CW, Kawas C, van Dyck CH, Sano M, Doody R, et al. Estrogen replacement therapy for treatment of mild to moderate Alzheimer disease: a randomized controlled trial. Alzheimer's Disease Cooperative Study. JAMA. 2000;283(8):1007-15.

155. Kawas C, Resnick S, Morrison A, Brookmeyer R, Corrada M, Zonderman A, et al. A prospective study of estrogen replacement therapy and the risk of developing Alzheimer's disease: the Baltimore Longitudinal Study of Aging. Neurology. 1997;48(6):1517-21.

156. Zandi PP, Carlson MC, Plassman BL, Welsh-Bohmer KA, Mayer LS, Steffens DC, et al. Hormone replacement therapy and incidence of Alzheimer disease in older women: the Cache County Study. JAMA. 2002;288(17):2123-9.

157. Shumaker SA, Legault C, Rapp SR, Thal L, Wallace RB, Ockene JK, et al. Estrogen plus progestin and the incidence of dementia and mild cognitive impairment in postmenopausal women: the Women's Health Initiative Memory Study: a randomized controlled trial. JAMA. 2003;289(20):2651-62.

158. Grodstein F, Colditz GA, Stampfer MJ. Postmenopausal hormone use and cholecystectomy in a large prospective study. Obstet Gynecol. 1994;83(1):5-11.

159. Chen Z, Bassford T, Green SB, Cauley JA, Jackson RD, LaCroix AZ, et al. Postmenopausal hormone therapy and body composition--a substudy of the estrogen plus progestin trial of the Women's Health Initiative. Am J Clin Nutr. 2005;82(3):651-6.

160. Margolis KL, Bonds DE, Rodabough RJ, Tinker L, Phillips LS, Allen C, et al. Effect of oestrogen plus progestin on the incidence of diabetes in postmenopausal women: results from the Women's Health Initiative Hormone Trial. Diabetologia. 2004;47(7):1175-87.

161. Vingerling JR, Dielemans I, Witteman JC, Hofman A, Grobbee DE, de Jong PT. Macular degeneration and early menopause: a case-control study. BMJ. 1995;310(6994):1570-1.

162. Klein BE, Klein R, Jensen SC, Ritter LL. Are sex hormones associated with age-related maculopathy in women? The Beaver Dam Eye Study. Trans Am Ophthalmol Soc. 1994;92:289-95; discussion 95-7.

163. Risk factors for neovascular age-related macular degeneration. The Eye Disease Case-Control Study Group. Arch Ophthalmol. 1992;110(12):1701-8.

164. Haapasaari KM, Raudaskoski T, Kallioinen M, Suvanto-Luukkonen E, Kauppila A, Laara E, et al. Systemic therapy with estrogen or estrogen with progestin has no effect on skin collagen in postmenopausal women. Maturitas. 1997;27(2):153-62.

165. Chen L, Dyson M, Rymer J, Bolton PA, Young SR. The use of high-frequency diagnostic ultrasound to investigate the effect of hormone replacement therapy on skin thickness. Skin Res Technol. 2001;7(2):95-7.

166. Ashcroft GS, Dodsworth J, van Boxtel E, Tarnuzzer RW, Horan MA, Schultz GS, et al. Estrogen accelerates cutaneous wound healing associated with an increase in TGF-beta1 levels. Nat Med. 1997;3(11):1209-15.

167. Grodstein F, Colditz GA, Stampfer MJ. Post-menopausal hormone use and tooth loss: a prospective study. J Am Dent Assoc. 1996;127(3):370-7, quiz 92.

168. Simon JA, Portman DJ, Kaunitz AM, Mekonnen H, Kazempour K, Bhaskar S, et al. Low-dose paroxetine 7.5 mg for menopausal vasomotor symptoms: two randomized controlled trials. Menopause. 2013;20(10):1027-35.

169. Simon JA, Portman DJ, Kazempour K, Mekonnen H, Bhaskar S, Lippman J. Safety Profile of Paroxetine 7.5 mg in Women With Moderate-to-Severe Vasomotor Symptoms. Obstet Gynecol. 2014;123 Suppl 1:132s-3s.

170. Lobo RA, Pinkerton JV, Gass ML, Dorin MH, Ronkin S, Pickar JH, et al. Evaluation of bazedoxifene/conjugated estrogens for the treatment of menopausal symptoms and effects on metabolic parameters and overall safety profile. Fertil Steril. 2009;92(3):1025-38.

171. Pinkerton JV, Harvey JA, Lindsay R, Pan K, Chines AA, Mirkin S, et al. Effects of bazedoxifene/conjugated estrogens on the endometrium and bone: a randomized trial. J Clin Endocrinol Metab. 2014;99(2):E189-98.

172. Archer DF, Lewis V, Carr BR, Olivier S, Pickar JH. Bazedoxifene/conjugated estrogens (BZA/CE): incidence of uterine bleeding in postmenopausal women. Fertil Steril. 2009;92(3):1039-44.

173. Portman D, Palacios S, Nappi RE, Mueck AO. Ospemifene, a non-oestrogen selective oestrogen receptor modulator for the treatment of vaginal dryness associated with postmenopausal vulvar and vaginal atrophy: a randomised, placebo-controlled, phase III trial. Maturitas. 2014;78(2):91-8.

174. Constantine G, Graham S, Portman DJ, Rosen RC, Kingsberg SA. Female sexual function improved with ospemifene in postmenopausal women with vulvar and vaginal atrophy: results of a randomized, placebo-controlled trial. Climacteric. 2014:1-7.

175. Archer DF, Carr BR, Pinkerton JV, Taylor HS, Constantine GD. Effects of ospemifene on the female reproductive and urinary tracts: translation from preclinical models into clinical evidence. Menopause. 2014.

176. Winther K, Rein E, Hedman C. Femal, a herbal remedy made from pollen extracts, reduces hot flushes and improves quality of life in menopausal women: a randomized, placebo-controlled, parallel study. Climacteric. 2005;8(2):162-70.

177. Estrogen and progestogen use in postmenopausal women: 2010 position statement of The North American Menopause Society. Menopause;17(2):242-55.

178. Santen RJ, Allred DC, Ardoin SP, Archer DF, Boyd N, Braunstein GD, et al. Postmenopausal hormone therapy: an Endocrine Society scientific statement. J Clin Endocrinol Metab;95(7 Suppl 1):s1-s66.

179. Canonico M, Oger E, Plu-Bureau G, Conard J, Meyer G, Levesque H, et al. Hormone therapy and venous thromboembolism among postmenopausal women: impact of the route of estrogen administration and progestogens: the ESTHER study. Circulation. 2007;115(7):840-5.

180. Renoux C, Dell'aniello S, Garbe E, Suissa S. Transdermal and oral hormone replacement therapy and the risk of stroke: a nested case-control study. BMJ;340:c2519.

181. Fournier A, Berrino F, Clavel-Chapelon F. Unequal risks for breast cancer associated with different hormone replacement therapies: results from the E3N cohort study. Breast Cancer Res Treat. 2008;107(1):103-11.

182. Warren MP. Historical perspectives in postmenopausal hormone therapy: defining the right dose and duration. Mayo Clin Proc. 2007;82(2):219-26.

183. Ockene JK, Barad DH, Cochrane BB, Larson JC, Gass M, Wassertheil-Smoller S, et al. Symptom experience after discontinuing use of estrogen plus progestin. JAMA. 2005;294(2):183-93.

The Non-Thyroidal Illness Syndrome

 

ABSTRACT
NTIS refers to a syndrome found in seriously ill or starving patients with low fT3, usually elevated RT3, normal  or low TSH, and if prolonged, low fT4. It is found  in a high proportion of patients in the ICU setting, and correlates with a poor prognosis if TT4 is <4ug/dl. The patho-physiology includes suppression of TRH release, reducedT3 and T4 turnover, reduction in liver generation of T3, increased formation of RT3, and tissue specific down-regulation of deiodinases, transporters, and TH receptors. Although long debated, tissue TH levels are definitely reduced, and tissue hypothyroidism is presumably present. This is often not clinically evident because of the brief duration, and reduced but not absent tissue levels of TH. Although recognized for nearly 4 decades, interpretation of the syndrome is contested, because of lack of data. Some observes, totally without data, argue that it is a protective response and should not be treated. Other observers (as in this review) present available data suggesting, but not proving, that thyroid hormone replacement is appropriate, not harmful, and may be beneficial. The best form of treatment (TRH,TSH,or T3+T4) and possible accompanying treatments (GHRH, Cortisol, nutrition, insulin) lack consensus. In this review current data are laid out for reader’s review and judgment.

 

DEFINITIONS

Serum thyroid hormone levels drop during starvation and illness. In mild illness, this involves only a decrease in serum triiodothyronine (T3) levels. However, as the severity and length of the illness increases, there is a drop in both serum T3 and thyroxine (T4). This decrease of serum thyroid hormone levels is seen in starvation, sepsis, surgery, myocardial infarction, bypass, bone marrow transplantation, and in fact probably any severe illness.1-9 The condition has been called the euthyroid sick syndrome (ESS). An alternative designation, which does not presume the metabolic status of the patient, is nonthyroidal illness syndrome, or NTIS. For more than 3 decades the interpretation of these changes has been debated   Many observers have considered the changes in hormone level to be laboratory artifacts, or if valid, not representative of true hypothyroidism, or if hypothyroidism was present, that it was a beneficial response designed to “spare calories” (1-21). More recently evidence has accumulated that central hypothyroidism, and altered peripheral metabolism of T4 and T3, combine to produce a state marked by diminished serum and tissue supplies of thyroid hormones. Nevertheless, some observers accept the low hormone levels as valid, but maintain that this is a (unique) situation in which such lack of hormone is not truly hypothyroidism (i.e., the “euthyroid sick syndrome”). Lastly, there is even greater uncertainly about hormone replacement therapy, in considerable  part because the opinion that replacement treatment should not be given has been repeated so many times, even though there is effectively no factual support for that view. We need controlled clinical trials in order to answer the question. It can not be solved by oft-stated opinions.

Low T3 States

Starvation, and more precisely carbohydrate deprivation, appears to rapidly inhibit deiodination of T4 to T3 by type 1 iodothyronine deiodinase in the liver, thus inhibiting generation of T3 and preventing metabolism of reverse T3 (rT3).10 Consequently there is a drop in serum T3 and elevation of reverse T3. Since starvation induces a decrease in basal metabolic rate,11 it has been argued, teleologically, that this decrease in thyroid hormone represents an adaptive response by the body to spare calories and protein by inducing some degree of hypothyroidism. Patients who have only a drop in serum T3, representing the mildest form of the NTIS, do not show clinical signs of hypothyroidism. Nor has it been shown that this decrease in serum T3 (in the absence of a drop in T4) has an adverse physiologic effect on the body or that it is associated with increased mortality.

Nonthyroidal Illness Syndrome With Low Serum T4

As the severity of illness, and often associated starvation, progresses, there is the gradual development of a more complex syndrome associated with low T3 and usually low T4 levels. Generally thyroid-stimulating hormone (TSH) levels are low or normal despite the low serum hormone levels, and rT3 levels are normal or elevated. A large proportion of patients in an intensive care unit setting have various degrees of severity of NTIS with low T3 and T4. Plikat et al. found that 23% of patients admitted to an ICU during a 2-year period had low free T3, low free T4, and low or normal TSH, and that these findings gave a greatly increased risk of death.12 Girvent et al. note that NTIS is highly prevalent in elderly patients with acute surgical problems and is associated with poor nutrition, higher sympathetic response, and worse postoperative outcome.13 Surprisingly, during the past 4 decades, some endocrinologists have assumed that NTIS is a beneficial physiologic response,14-17 but factual evidence for this view is unavailable. However it seems illogical to consider  NTIS as an evolutionarily derived  physiologic response, since survival with the severity of illness seen in NTIS patients would be almost impossible except in modern ICUs.

A marked decrease in serum T3 and T4 in NTIS is associated with a high probability of death. NTIS was found in a group of 20 patients with severe trauma, among whom 5 died, and the drop in T3 correlated with the Apache II score.18 NTIS found in patients undergoing bone marrow transplantation was associated with a high probability of fatal outcome.19 NTIS was typical in elderly patients undergoing acute surgery and was associated with a worse prognosis.20 All of 45 non-dopamine-treated children with meningococcal septicemia had low T3, T4, and thyroxine-binding globulin (TBG), without elevated TSH. When serum T4 levels drop below 4 g/dL, the probability of death is about 50%, and with serum T4 levels below 2 g/dL, the probability of death reaches 80%.21-23 Obviously such associations do not prove that hypothyroidism is the cause of these complications or deaths, but the fact of hypothyroidism must at least raise the consideration of treatment.

Interpretations of NTIS

Several conceptual explanations of NTIS can be followed through the literature:

1.         The abnormalities represent test artifacts, and assays would indicate euthyroidism if proper tests were employed.

2.         The serum thyroid hormone abnormalities are due to inhibitors of T4 binding to proteins, and the tests do not appropriately reflect free hormone levels. Proponents of this concept may or may not take the position that a binding inhibitor is present throughout body tissues, rather than simply in serum, and that the binding inhibitor may also inhibit uptake of hormone by cells or prevent binding to nuclear T3 receptors and thus inhibit action of hormone.

3.         In NTIS, T3 levels in the pituitary are normal because of enhanced local deiodination. In this concept the pituitary is actually euthyroid, while the rest of the body is hypothyroid. This presupposes enhanced intrapituitary T4 > T3 deiodination as the cause.

4.         Serum hormone levels are in fact low, and the patients are biochemically hypothyroid, but this is (teleologically) a beneficial physiologic response and should not be altered by treatment.

5.         Lastly, NTIS is in part a form of secondary hypothyroidism, the patient’s serum and tissue hormone levels are truly low, tissue hypothyroidism is present, this is probably disadvantageous to the patient, and therapy should be initiated if serum thyroxine levels are depressed below the danger level of 4 μg/dL.

 

SERUM HORMONE LEVELS AND TISSUE HORMONE SUPPLIES IN NTIS

Serum T3 and Free T3

With few exceptions, reports on NTIS indicate that serum T3 and free T3 levels are low.24-30

Liver Iodothyronine D1 normally produces up to 80% of circulating T3 via T4>T3 deiodination, the remainder coming from the thyroid directly, or by a contribution from ID2 in muscle as noted below. ID1 in liver is down-regulated in severe illness, and this is certainly an important contributor to the low T3 in blood. One presumed cause is reduced nutrition, especially of carbohydrate, but direct effects of cytokines on liver may also be involved  The problem presumably is exacerbated by hypothyroidism, which also down-regulates ID1.

Chopra and co-workers reported that free T3 levels were low (Fig. 1),31 or in a second report, often normal.32 However, it is important to note that in the second report, the patients with “NTIS” actually had average serum T4 levels that were above the normal mean and did not have significant NTIS. Sapin et al. compared free T3 levels found in patients with NTIS by direct dialysis, microchromatography, analogue, two-step immune extraction, and a labeled antibody RIA method.30 Results were significantly below normal by five of the methods and low in the most severe cases by one method. Faber et al. evaluated thyroid hormone levels in 34 seriously ill patients, most of whom had low T4 and free T4 index values, and found generally normal free T3 and free T4 using an ultrafiltration technique.33 A point to consider is that some ultrafiltration techniques fail to exclude thyroid hormone–binding proteins from the filtrate and give spuriously high free hormone values.34

Figure 1. Free T3 concentrations in different groups of patients, as reported by Chopra et al, reference 32. In this report, patients with NTIS have significantly lowered Free T3 levels than do normal subjects.

Figure 1. Free T3 concentrations in different groups of patients, as reported by Chopra et al, reference 32. In this report, patients with NTIS have significantly lowered Free T3 levels than do normal subjects.

Serum rT3 may be reduced, normal, or elevated and is not a reliable indicator of abnormal thyroid hormone supply. While it may be expected that rT3 should always be elevated, this is not true, and often it is within the normal range. Peeters et al.35 found in patients with NTIS, serum TSH, T4, T3, and the T3/rT3 ratio were lower, whereas serum rT3 was higher than in normal subjects (P < 0.0001). Liver D1 is down-regulated, and D3 (which is not evident in liver and skeletal muscle of healthy individuals) is induced, particularly in disease states associated with poor tissue perfusion. The level of rT3 reflects the action of several enzymes and presumably, as well, tissue metabolic function. Induction of D3 would tend to increase rT3. Degradation of rT3 is reduced by decreased function of the same D1 enzyme that generates T3. However, formation of rT3 is limited by the low level of substrate (T4) in serum and in tissues and perhaps by inhibition of T4 entry into cells. Personal experience treating patients with NTIS (unpublished) shows that when T4 is given and repletes serum hormone levels, generation of rT3 rapidly increases, and levels often become significantly elevated.

Serum T4

Serum T4 levels are reduced in NTIS in proportion to the severity and, probably, length of the illness.24-35 In acute, short-term trauma such as cardiac bypass36 or in short-term starvation,37 there is no drop in serum T4. However, with increasing severity of trauma, illness, or infection, there is a drop in T4 which may become extreme. As indicated, serum T4 levels below 4 μg/dL are associated with a marked increased risk of death (up to 50%), and once T4 is below 2, prognosis becomes extremely guarded. In neonates, low total T4 and TSH are associated with a greater risk of death and severe intraventricular hemorrhage. It is suggested that thyroid hormone supplementation might be a potential benefit in infants with the lowest T4 values.27

Total serum T4 is reduced in part because of a reduction in TBG. One reason for this reduction appears to be because of cleavage of TBG. Schussler’s group recognized a rapid drop in TBG to 60% of baseline within 12 hours after bypass surgery, and their data suggest that this is due to cleavage of TBG by protease, which causes TBG to lose its T4-binding activity.38 Further studies by this group demonstrated the presence of a cleaved form of TBG present in serum of patients with sepsis.39

The impact of meningococcal sepsis on peripheral thyroid hormone metabolism and binding proteins was studied in 69 children with meningococcal sepsis. All children had decreased total T3 and total T3/rT3 ratios without elevated TSH. Lower total T4 levels were related to increased turnover of TBG by elastase. Lowered TBG is a partial explanation for lower total T4 and T3 in NTIS.40

Serum Free Thyroxine

A major problem in understanding NTIS is in analyzing data on the level of free T4. Free T4 is believed by most workers to represent hormone availability to tissues, although it is in fact intracellular T3 that binds to the receptors. The results of free T4 assays in NTIS are definitely method dependent. They may be influenced by a variety of variables, including (alleged) inhibitors present in serum or the effect of agents such as drugs, metabolites, or free fatty acids in the serum or assay. Assays which include an estimate of TBG capacity to estimate free hormone typically return low values for calculated free thyroxine in NTIS. Methods using T3 analogs in the assay also give levels that are depressed. The free T4 level determined by dialysis varies widely, as does T4 measured by ultrafiltration25-29; the majority of reports are of low values, but in some samples nnormal or rarely elevated values.25,26,41-43

In theory, methods utilizing equilibrium dialysis may allow dilution of dialyzable inhibitors. Compounds such as 3-carboxy-4-methyl-5-propyl-2-furan-propanoic acid, indoxyl sulfate, and hippuric acid, can accumulate in severe renal failure.44 However, these compounds probably do not interfere with serum hormone assays. Free fatty acids, if elevated to 2 to 5 mmol/L, can displace T4 binding to TBG and elevate free T4. Free fatty acids almost never reach such levels in vivo.45,46 However, even small quantities of heparin (0.08 units/kg given IV, or 5000 units given SC), commonly given to patients in an ICU, can lead to in vitro generation of free fatty acids during extended serum dialysis for “free T4“ assay and falsely augment apparent free hormone levels.47 This is probably a common and serious problem, which explains many instances of apparently elevated free T4 levels in patients with acute illness.

Results obtained using ultrafiltration also are variable. Wang et al.48 found that in patients with NTIS, free T4 measured by ultrafiltration was uniformly low (average of 11.7 ng/L), but when measured by equilibrium dialysis, free T4 was near normal, at 18 ng/L. By ultrafiltration, free T3 was also (not surprisingly) found to be low and similar to free T3 by radioimmune assay. Chopra32 found levels below the normal mean, ±2 SD, when measured by dialysis; 6 of 9 were low when measured by ultrafiltration, and 7of 9 were low when measured by standard resin-uptake-corrected free T4. The means of the NTIS patients in this study were clearly below the mean of normals.

Thus, although free T4 is low in most assays that involve a correction for TBG levels, there is still some question as to the true free T4 in patients with NTIS. It is of interest that this problem does not carry over to estimates of free T3, which are depressed in most studies. There might be two reasons for this difference. Firstly, the depression of total T3 is proportionately greater than of total T4. Secondly, factors which affect thyroid hormone binding are more apt to alter T4 assays than T3, since T4 is normally more tightly bound to TBG than is T3.

 

IS THERE EVIDENCE FOR SUBSTANCES IN SERUM WHICH CAN AFFECT T4 BINDING TO PROTEINS?

Mendel et al.49 carefully review the studies that have claimed the presence of dialyzable inhibitors of binding and point out that many of these studies must be viewed with caution44,45,50-53 .Numerous artifacts are present in both dialysis assays and ultrafiltration assays. They also point out that while the low free T4 by resin uptake assays found in NTIS generally do not agree with the clinical status of the patient, it is equally true that clinical assessment generally does not fit with the high free T4 results found by some equilibrium dialysis assays in NTIS.

An argument that completely refutes the importance of factors in serum inhibiting binding of thyroid hormone is provided in the clinical study of Brent and Hershman (Fig. 2).54 These

Figure 2. Patients with severe NTIS were randomized and left untreated or given T4 iv over two weeks. Serum T3, T4, and TSH concentrations are shown for the survivors of the control filled circles), and T4-treated empty circles), groups during the study period and at the time of follow-up. Upper and lower lines designate the normal range. Note the prompt recovery of T4 values to the normal range immediately following i.v. treatment with T4. Also note the elevated TSH levels in some patients. T3 levels did not return to normal following T4 treatment for up to two weeks. (Reference 54)

Figure 2. Patients with severe NTIS were randomized and left untreated or given T4 iv over two weeks. Serum T3, T4, and TSH concentrations are shown for the survivors of the control filled circles), and T4-treated empty circles), groups during the study period and at the time of follow-up. Upper and lower lines designate the normal range. Note the prompt recovery of T4 values to the normal range immediately following i.v. treatment with T4. Also note the elevated TSH levels in some patients. T3 levels did not return to normal following T4 treatment for up to two weeks. (Reference 54)

researchers gave 1.5 μg of T4 per kg body weight daily to 12 of 24 patients with severe NTIS and followed serum hormone levels over 14 days. T4 levels returned to the normal range within 3 days of therapy. Thus the serum thyroxine pool was easily replenished, and T4 levels reached normal values. Not surprisingly, because of reduced T4>T3 deiodination, T3 levels did not return to the normal range until the end of the study period in the few patients who survived. However, the ability of intravenous thyroxine in replacement doses to promptly restore the plasma pool to normal clearly shows that neither a loss of serum TBG nor an inhibitor of binding could be the main cause of low serum T4 in this group of severely ill patients.

With growing acceptance of decreased thyroid secretion and decreased peripheral t3 production as causes of low T4 and T3, there has been little emphasis on serum T4 binding inhibitors in recent literature. Some contribution by low TBG levels may, or may not (see below) play a role, but any role for binding inhibitors in producing this syndrome must be marginal

TSH LEVELS

Serum TSH in NTIS is typically normal or reduced and may be markedly low, although usually not less than 0.05 μU/mL.16,24,25,28,29,31,55 However, to use usual endocrinology logic, these TSH levels are almost always inappropriately low for the observed serum T4 and T3. Third-generation assays with sensitivity down to 0.001 U/mL may allow differentiation of patients with hyperthyroidism from those with NTIS, although there can be overlap in these very disparate conditions.56 Serum TSH in patients with NTIS may have reduced biological activity, perhaps because of reduced thyrotropin-releasing hormone (TRH) secretion and reduced glycosylation. Some patients are found with a TSH level above normal, and elevation of TSH above normal commonly occurs transiently if patients recover from NTIS (Fig. 3).29,54 This elevation of TSH strongly suggests that the patients are recovering from a hypothyroid state, during which the ability of the pituitary to respond had been temporarily inhibited.

Figure 3. T3 and TSH concentrations are shown in patients with nonthyroidal illness who were eventually discharged from hospital (left panels). The broken line indicates ± 2 SD of the mean value in the normal subjects. The right panel displays T3 and TSH concentrations in patients with NTIS who died. Subjects are indicated by numbers. Note the elevated TSH in some patients who recovered, and the generally dropping T3 and low TSH levels in patients who died. (Reference 29)

Figure 3. T3 and TSH concentrations are shown in patients with nonthyroidal illness who were eventually discharged from hospital (left panels). The broken line indicates ± 2 SD of the mean value in the normal subjects. The right panel displays T3 and TSH concentrations in patients with NTIS who died. Subjects are indicated by numbers. Note the elevated TSH in some patients who recovered, and the generally dropping T3 and low TSH levels in patients who died. (Reference 29)

 

Responsiveness of the pituitary to TRH during NTIS is variable: many patients respond less than normal,57 and others respond normally.58 “Normal” responsiveness in the presence of low TSH may suggest that there is a hypothalamic abnormality as a cause of the low TSH and low T4. There is also a diminution or loss of the diurnal rhythm of TSH,59 and in some studies, there is evidence for reduction of TSH glycosylation, with lower TSH bioactivity.60 A logical explanation is that the low TSH is in fact the proximate cause of the low thyroid hormone levels. Hypothalamic function is impaired in patients with NTIS and TRH mRNA is low,  resulting in low TSH and thus low output of thyroid hormones by the thyroid.

There is other evidence of diminished hypothalamic function in patients with serious illness. Serum testosterone drops rapidly, as do follicle-stimulating hormone (FSH) and luteinizing hormone (LH).61,62 Typically serum cortisol is elevated as part of a stress response, and because metablism of corticol is reduced. Some patients develop hypotension in association with apparent transient central hypoadrenalism, have low or normal serum ACTH, and cortisol levels under 20 μg/dL. Some of these patients respond dramatically to cortisol replacement and may manifest normal adrenal function at a later time if they recover.

Centrally mediated hyposomatotropism, hypothyroidism, and pronounced hypoandrogenism were observed in a study of patients in the catabolic state of critical illness. In these patients, pulsatile LH secretion and mean LH secretions are very low, even in the presence of extremely low circulating total testosterone and low estradiol. Pulsatile growth hormone (GH) and TSH secretion are also suppressed. Interleukin 1 β (IL-1β) levels are normal, whereas IL-6 and tumor necrosis factor α (TNF-α) are elevated. Exogenous IV gonadotropin-releasing hormone (GnRH) partially return serum testosterone levels toward normal but do not completely overcome hypoandrogenism, suggesting that combined deficiency of GH, GnRH, and TSH secretagogues may be important in this low androgen syndrome.63

THYROID HORMONE TURNOVER

Kaptein et al.64,65 studied a group of patients who were critically ill, all of whom had total T4 below 4 μg/dL, low fT4 index, low normal free T4 by dialysis, and TSH which was normal or slightly elevated. In these patients, the mean T4 by dialysis was significantly below the normal mean. There was on average a 35% decrease in thyroxine disposal per day (Table 1). The T4 production rate in NTIS was significantly below the mean of 17 normal subjects (p < 0.005). In a similar study of T3 kinetics,65 free T3 was found to be 50% of normal serum values. The production rate of T3 was reduced by 83% (Table 2). These two studies document a dramatic reduction in provision of T4 and T3 to peripheral tissues, which would logically indicate that the effects of hormone lack (hypothyroidism) should be present. A third study reported dramatically reduced total T4 and T3 turnover, with normal thyroidal secretion of T3 in patients with NTIS due to uremia.66 However, this was a calculated rather than directly measured value of T3 secretion, was highly variable, and does not negate the extreme reduction in T3 supply due to diminished T4 >T3 conversion in peripheral organs.

T4 ENTRY INTO CELLS AND GENERATION OF T3

Thyroid hormone is transported actively into tissues by several specific transporters including MCT8, and in the pituitary OATP1C1. In the cell it is metabolized by enzymes which activate it to T3, or inactivate it to rT3, or promote excretion via sulfation or glucuronidation. Iodotyrosine deiodinase type 1 (ID1) is found in liver, kidney and thyroid, and the enzyme present in liver is considered a main source of T3, possibly providing 80% of the total, the remainder coming largely from the thyroid. ID1 is down-regulated in hypothyroidism, and in NTIS, reducing serum T3 levels.  ID2 is present in brain and pituitary, and is responsible for local production of T3 in those tissues. Recent data show that D2 present in muscle may also contribute to serum T3. ID2 is up-regulated by hypothyroidism, and is up-regulated in  NTIS. The third enzyme, ID3, deiodinates the inner thyronine ring, converting T4 to rT3 and T3 to T2. It’s activity in liver is up-regulated in NTIS.

Using deiodination of T4 as an index of cellular transport of T4 into rat hepatocytes, Lim et al.67 and Vos et al.68 found that serum from patients with NTIS inhibited T4 uptake. Sera from critically ill NTIS patients caused reduced T4 uptake compared to control sera in one study, and the authors considered elevated nonesterified fatty acids (NEFA) and bilirubin and reduced albumin to play a role. Serum from patients with mild NTIS did not cause impaired deiodination of T4 and T3.69 Inhibition of uptake of T4 into hepatocytes caused by sera of patients with NTIS also was observed by Sarne and Refetoff.70 There is a diminution in the “reducing equivalents” available for the deiodination of T4 to T3 in liver, and presumably elsewhere, thus lowering transport and the function of the type 1 iodothyronine deiodinase.71 In animals, and probably in man, there is also a drop in the level of type 1 iodothyronine deiodinase enzyme, apparently due to hypothyroidism, since it can be reversed by giving T3. Recently a study was performed on blood, liver, and skeletal-muscle biopsies of patients immediately after death in intensive care unit settings. Liver T4 deiodinase 1 was found to be down-regulated, and deiodinase 3 was induced in liver and muscle, especially in situations associated with poor tissue perfusion. These changes contribute to the low generation of T3 and its increased metabolism in NTIS, thus lowering the intracellular T3 levels.35

Table 1. T4 Kinetics in the Low T4 State of Nonthyroidal Illness64

Case Number

TT4 (µg/dL)

FT4 (ng/dL)

PR (µg/d/m2)

Normal Subjects (n = 19)

     

Mean

7.1

2.21

50.3

±SE

0.4

0.13

3.4

Sick Patients

     

1

2.7

2.05

32.4

2

3.0

1.23

51.1

3

1.2

0.48

39.0

4

1.4

1.04

23.7

5

1.3

0.75

22.2

6

3.0

1.35

34.6

7

1.9

1.33

36.6

8

2.0

1.88

25.3

9*

0.4

0.28

10.0

10*

1.5

1.50

13.7

11*

1.6

1.70

18.4

Mean

1.8

1.24

27.9

±SE

0.2

0.17

3.7

P

<0.001

<0.001

<0.001

FT4, Free thyroxine; PR, production rate; TT4, total thyroxine.

*Patients receiving dopamine.

All P values are for unpaired t tests.

Table 2. T3 Kinetics in the Low-T4 State of Nonthyroidal Illness65

Case Number

TT3 (ng/dL)

FT3 (pg/dL)

PR (µg/d/m2)

Normal Subjects (n = 12)

     

Mean

162

503

23.47

±SE

5

46

2.12

Sick Patients

     

3

30

272

6.18

5

42

247

5.67

6

25

151

5.41

7

34

266

8.39

12*

45

282

6.07

Mean

35

244

6.34

±SE

4

24

0.53

P

<0.001

<0.001

<0.005

FT3, Free triiodothyronine; PR, production rate; TT3, total triiodothyronine. *Patient receiving dopamine.

In theory, reduced cellular uptake (acting  alone) would cause tissue hypothyroidism, reduced T3 generation and serum T3 levels, and elevated serum T4, which is not observed. It is likely that reduced hormone supply in NTIS is caused by multiple factors, and that reduced cell uptake, if present, is one of the factors. T4 is converted to T3, although at a reduced rate. In addition, T4 is rapidly converted to rT3 by an intracellular process, suggesting that entry into cells is not seriously impaired, but the pathways of intracellular deiodination are abnormal.

 

THYROID HORMONE IN TISSUES

There are increasing data on thyroid hormone in tissues of patients with NTIS.72 In one study, there was of a dramatically reduced level of T3 in tissues (Table 3). While most samples had very low levels of T3 compared to normal tissues, some patients with NTIS showed sporadically and inexplicably high levels of T3 in certain tissues, especially skeletal muscle and heart.

Table 3. Tissue T3 Concentrations in Nonthyroidal Illness Syndrome (nmol of T3/kg of Wet Weight)72

 

Control Group

NTI Group

Tissue

Mean

SD

P

Mean

SD

Cerebral cortex

2.2

0.9

<.05

1.2

1.1

Hypothalamus

3.9

2.2

<.01

1.4

1.2

Anterior pituitary

6.8

2.5

<.005

3.7

1.1

Liver

3.7

2.3

<.01

0.9

0.9

Kidney

12.9

4.3

<.001

3.7

2.8

Lung

1.8

0.8

<.01

0.8

0.5

Skeletal muscle

2.3

1.2

NS

> 10.9

 

Heart

4.5

1.5

NS

> 16.3

 

NS, Not significantly different; NTI, nonthyroidal illness; T3, triiodothyronine.*Patients receiving dopamine.

Peeters et al.73 investigated 79 patients who died after intensive care, some of whom received thyroid hormone treatment. Tissue iodothyronine levels were positively correlated with serum levels, indicating that the decrease in serum T3 during illness is associated with decreased levels of tissue T3. Higher serum T3 levels in patients who received thyroid hormone treatment were accompanied by higher levels of liver and muscle T3, with evidence for tissue-specific regulation. Tissue rT3 and the T3/rT3 ratio were correlated with tissue deiodinase activities. Monocarboxylate transporter 8 expression was not related to the ratio of the serum/tissue concentrations of the different iodothyronines.73

TR LEVELS

Information on expression of TRs in human tissues during illness is limited. Increased expression of the messenger ribonucleic acid (mRNA) for thyroid hormone receptors α1, α2, and β1 has been reported in cardiac tissue of patients with dilated cardiomyopathy; α1 and α2 isoforms also had increased expression in ischemic heart disease.74 Rodriguez-Perez et al. studied subcutaneous fat and skeletal muscle in patients with septic shock.75 In muscle, mRNA for TRβl and RXR gamma was reduced, and mRNA for RXR alpha was increased, compared to normals. In adipose tissue, MCT8, TRβ1, TRα1, and RXR gamma mRNAs were lower. The authors conclude that in these patients, tissue responses were related to decreased hormone levels and decreased hormone action. In animals, starvation and illness are associated with a reduction in thyroid hormone receptor levels. In experimental studies in mice, LPS induces NTIS, and this is associated with an early decrease in binding of the RXR/TR dimer to DNA due to limiting amounts of RXR, and later an up to 50% decrease in levels of RXR and TR protein.- Lado-Abeal  and co workers found in humans with prolonged NTIS that expression of TRbeta1, TRalpha1, and RXRgamma in striated muscle were reduced compared to normals, and that these changes were unrelated to expression of NFkB1(Figure 4)(78). .

Figure 4 . THRA, THRB1, RXRG, RXRA, AND PPARG PROTEIN levels were evaluated by western blotting.

Figure 4 . THRA, THRB1, RXRG, RXRA, AND PPARG PROTEIN levels were evaluated by western blotting.

 

 

 

ORGAN SPECIFIC RESPONSES IN NTIS

In contrast to a uniform whole body response, there are wide variations in responses to thyroid hormone supply and action in different tissues, and between the response to acute illness and chronic illness. These multifactorial systems involve serum TH levels, TH transporters, deiodinases, TH receptors, and enzyme responses which are under different regulation in individual tissues (79). In reviewing these data it is useful to note that meaning of data often must be “interpreted”, and that this can depend on the mind-set of the reviewer. In the hypothalamus in an animal model of NTIS, TH transporters MCT10 and OATP1C1 (but not MCT8) were increased, and hypothalamic D2, considered a major source of local T3, was up-regulated, but there was no corresponding increase in tissue T3..  TRalpha and beta mRNA expression  levels were not altered (Figure 4)(80). Tissue levels of T3 and T4 are reduced in chronic NTIS in both experimental models and in humans. Thus the know low TRH mRNA levels in the hypothalamus reflect the action of neural signals, and not an hypothesized local tissue hyperthyroidism. On the other hand, the “low normal” T3 levels in the hypothalamus, in the presence of low serum T3 levels, presumably reflect the actionof D2, and may provide a partial explanation of why TSH is not usually elevated in NTIS. Data on the pituitary are few. D2 levels have not shown consistent changes. In animal models TRbeta2 levels are reported to be reduced in acute NTIS. In humans with fatal illness, pituitary T3 levels were low. The overall picture is of central down-regulation of the hypothalamus and pituitary and low levels of tissue T3 despite increments in transporter activity and D2 deiodinase(Figure 5). These findings fit with the observed correction of TSH and TH levels in human NTIS through administration of TRH.

Figure 5 . T3 and T4 content in Hypothalamus of chronically ill animals with NTIS.(From Mebis et al(80).

Figure 5 . T3 and T4 content in Hypothalamus of chronically ill animals with NTIS.(From Mebis et al(80).

Liver D1 and D3 activity are reduced in acute NTIS. In man MCT8 and MCT10 may be reduced in acute but not chronic NTIS. Several enzymes that are responsive to TH have reduced activity in acute NTIS in animals. The data fit with reduced metabolic activity in this organ. In chronic  NTIS in man, reduced  serum T3 and T4 and normal or elevated RT3  are characteristic. Liver T3 is low in chronic human NTIS, and is directly related to serum T3. MCT8, but not MCTt10, are increased in liver and muscle in prolonged NTIS (81). Expression levels of TRalpha and beta mRNA are reported to be reduced, or increased. Metabolic activity is probably reduced, but in relation to oxygenation and nutrients as well as TH activity. In a rabbit model of chronic NTIS, serum and liver T3  and liver D1 activity were low. Interestingly replacement with basal levels T4 or T3 did not reverse these abnormalities, but 3-5 fold increments, or TRH administration, did so (82). This study offers several interesting points. There was a very strong correlation between serum  and tissue hormone levels, so tissue entry was snot a problem. The requirement for more than replacement dose T4 to restore tissue hormone is mainly due to repletion of very diminished stores of T4, in this 6 day treatment  protocol. No evidence was found for an important role of T4-sulfo  conjugates. The response in muscle is less well defined. D2 increases and D3 is decreased in acute NTIS in an animal model, and there is evidence for decreased TR expression. Changes in enzyme responses do not show a consistent pattern.  In chronic NTIS, human muscle D3 is augmented,  while D2 has been found low, or increased. Muscle MCT8 is increased (81), and this has been proposed as a response to hypothyroidism, since TH treatment in a rabbit model of NTIS returns the transporters to normal. Enzymatic activity is presumed low but good data are lacking.  Prolonged infusion of lipopolysaccharide in pigs induced a severe NTIS state, associated with generally low tissue TH levels, reduced TH transporters, and low TR-beta levels, suggesting reduced TH sensitivity and hypothyroidism(83).

 

ARE PATIENTS WITH NTIS CLINICALLY HYPOTHYROID?

It is straightforward that the typical clinical parameters of severe hypothyroidism are absent in patients with NTIS. However, these patients usually present with a serious illness and are diagnostically challenging in view of their complicated state. Many are febrile, have extensive edema, have sepsis or pneumonia, may have hyper metabolism associated with burns, have severe cardiac or pulmonary disease, and in general have features that could easily mask evidence of hypothyroidism. Further, the common clinical picture of hypothyroidism does not develop within 2 to 3 weeks of complete thyroid hormone deprivation, but rather requires a much longer period for expression. General laboratory tests are also suspect. Thus starvation or disease-induced alterations in cholesterol, liver enzymes, TBG, creatine kinase, and even basal metabolic rate generally rule out the use of these associated markers for evidence of hypothyroidism. Angiotensin-converting enzyme levels are low,84 as seen in hypothyroidism, while high-affinity testosterone-binding globulin (TeBG) and osteocalcin levels are not altered.85Antithrombin III levels are reduced in a septic rat model of NTIS. T3 supplementation returned the sepsis-induced decrease in antithrombin III levels toward normal.86

 

MECHANISM OF THYROID HORMONE SUPPRESSION IN NTIS

It is probable that the cause of NTIS is multifactorial and may differ in different groups of patients. Specifically, the changes in liver disease and renal disease are probably somewhat different from those occurring in other forms of illness. Certainly one important cause of the drop in serum T3 is a decreased generation of T3 by type 1 iodothyronine deiodinase.87 Reduced entry of T4 into cells is not a major problem. Some studies have suggested that individuals with NTIS may have selenium deficiency, and this may contribute to a malfunction of the selenium-dependent iodothyronine deiodinase.However, supplements of 500 μg of selenium given to patients in a surgical ICU during the first 5 days after serious injury caused only modest changes in thyroid hormones. The data did not suggest a major role for selenium deficiency in this condition.88

The overall daily metabolic consumption of thyroid hormone, both thyroxine and T3, is radically diminished in the NTIS syndrome in the presence of low hormone serum levels. The reduced degradation cannot produce the lowering of serum hormone levels; a primary reduction in degradation would increase serum hormone. The change in degradation must be due to the low hormone supply, and other factors. Schussler and co-workers have observed a sharp drop in TBG levels during cardiac bypass surgery, which their studies indicate is due to some selective consumption of TBG. It is possible that this occurs because of activation of serine protease inhibitors (serpins) at sites of inflammation, which cleave the TBG into an inactive form.38

Considerable evidence suggests that an alteration in hypothalamic and pituitary function causes the low production of T4, which in turn causes the low production of T3. In rats, starvation reduces hypothalamic mRNA for TRH, reduces portal serum TRH, and lowers pituitary TSH content.89A recent study documents low TRH mRNA in hypothalamic paraventricular nuclei90 in NTIS patients (Fig. 6). Responses to administered TRH vary in different reports, being suppressed or even augmented.57,58 Administration of TRH has been suggested as an effective

 Figure 6. In situ hybridization study demonstrating mRNA for TRH in the periventricular nuclei of a subject who died with NTIS in Panel A, and a subject who died accidentally in Panel B. mRNA for TRH is significantly reduced in patients with NTIS. (Reference 90)

Figure 6. In situ hybridization study demonstrating mRNA for TRH in the periventricular nuclei of a subject who died with NTIS in Panel A, and a subject who died accidentally in Panel B. mRNA for TRH is significantly reduced in patients with NTIS. (Reference 90)

means of restoring serum hormone levels to normal in individuals with NTIS. A recent report by Van den Berghe and co-workers proves that administration of TRH to patients with severe NTIS leads directly to increased TSH levels, increased T4 levels, and increased T3 levels.91 This data is strong support (albeit not proof) for the role of diminished hypothalamic function as a crucial factor in NTIS.

Quite possibly the production of TRH, and responses to TRH, are reduced by cytokines (to be discussed later) or by glucocorticoids.92 The diurnal variation in glucocorticoid levels at least in part controls the normal diurnal variation in TSH levels, perhaps by affecting pituitary responsiveness to TRH.93 High levels of glucocorticoids in Cushing’s disease suppress TSH and cause a modest reduction in serum hormone levels.94 High levels of glucocorticoids are known to suppress pituitary response to TRH in man.92Stress-related elevation of glucocorticoids in animals causes suppression of TSH and serum T4 and T3 hormone levels.95 Thus stress-induced glucocorticoid elevation may be one factor affecting TRH and TSH production.

Why should pituitary production of TSH be diminished in the presence of low serum thyroid hormone levels? One idea was that augmented intrapituitary conversion of T4 to T3 allowed the pituitary to remain suppressed while the rest of the body was actually hypothyroid. While some data supported  this idea in a uremic rat model of NTIS96, careful studies in both experimental  animals(82,83) and man, described above, disprove this concept.

 Another suggestion is that some other metabolite of thyroxine may be involved in control of pituitary responsiveness. For example, possibly triiodothyroacetic acid (triac) or tetraiodothyroacetic acid (tetrac) generated by metabolism of thyroxine could control pituitary responsiveness,92 but there is no experimental proof of this idea, and even if true, it would mean that the pituitary was normal but the rest of the body hypothyroid. As suggested earlier, elevated serum cortisol levels could play a role. The most obvious possibility is that low TSH stems from diminished TRH production, as previously described. It must also be remembered that the defect in pituitary function is not restricted to TSH, but that LH and FSH are also suppressed in seriously ill patients, and testosterone is reduced, in contrast to the generally augmented glucocorticoid levels. Quite possibly these changes are the effect on the hypothalamus of neural integration of multiple factors including stress, starvation, glucocorticoids, and cytokines.

Van den Berghe has stressed that the changes in endocrine function seen during severe illness have a biphasic course. Possibly the initial suppression of T3 levels represents a genetically engineered adaptive response of the organism, allowing reduced metabolic rate and conservation of energy and protein stores for a longer period of time, while the animal or man goes through a period of starvation. However, the circumstances surrounding severe illness, and the resuscitative efforts applied in an intensive care unit over 1 or more weeks, seem to be a different reponse. This second phase of the syndrome, with associated suppression of thyroid hormone and other pituitary hormones and a variety of other changes, may represents a maladaptive response. Patients in this situation tend to have elevated insulin levels, nitrogen wasting, retention of fats if calories are made available, and a variety of other metabolic abnormalities that include neuropathy and cardiomyopathy. These authors consider that provision of multiple hormonal support, including thyroid hormone, growth hormone, and androgens, may be beneficial.97,98

 

CYTOKINES IN NTIS

In a series of septic patients studied shortly after admission to an ICU, total T4, free T4, total T3, and TSH were depressed, and IL-1β, soluble interleukin-2 receptor (sIL-2R), IL-6, and TNF-α were elevated.99 The hypothalamo-pituitary-adrenal axis was activated as expected. The data suggest central suppression of TSH as the cause of the problem, but the relation to cytokines is unclear, as seen in the following reports. Hermus et al.100 showed that continuous infusion of IL-1 in rats causes suppression of TSH, T3, and free T4. Higher doses of IL-1 were accompanied by a febrile reaction and suppression of food intake, which presumably played some role in the altered thyroid hormone economy. IL-1 did not reproduce the diminution in hepatic 5′-deiodinase activity believed to be so characteristic of NTIS. IL-1 is also known to impair thyroid hormone synthesis by human thyrocytes and is enhanced in many diseases associated with NTIS.101 van der Poll et al.102 studied the effect of IL-1 receptor blockade in human volunteers to determine if it could alter the NTIS induced by endotoxin. Blockade of IL-1 activity was achieved by infusing recombinant human IL-1 receptor antagonist, but this did not prevent the drop in T4, free T4, T3, and TSH or the rise in rT3 caused by endotoxin. This is evidence against an important role for IL-1.

Interferon γ

Interferon-γ (IFN-γ) 100 μg/m2 administered subcutaneously to normal volunteers did not alter TNF-α levels, caused a small elevation of IL-6 levels, and thus did not support a role for IFN-γ in the pathogenesis of the euthyroid sick syndrome in humans.103

Tumor Necrosis Factor

TNF is another proinflammatory cytokine that is thought to be involved in many of the illnesses associated with NTIS.Infusion of recombinant TNF in man by van der Poll et al.104 produced a decrease in serum T3 and TSH and an increase in rT3. Free T4 was transiently elevated in association with a significant rise in FFA levels. These studies suggest that TNF could be involved in producing NTIS. Recombinant IL-6 given to humans activates the hypothalamic pituitary-adrenal axis, and this could play a role in suppressing TSH production. However, Chopra et al.105 did not find TNF to be closely correlated with hormone changes in NTIS. van der Poll et al.1063 gave human subjects endotoxin, which caused lowering of T4, free T4, T3, and TSH. TNF blockade by a recombinant TNF receptor-IgG fusion protein did not alter the response, indicating that TNF did not cause the changes in hormone economy induced by administration of endotoxin. Nagaya et al.107 proposed a mechanism through which TNF could reduce serum T3. TNF-α was found during in vitro studies to activate nuclear factor kB (NF-kB), which in turn inhibits the T3-induced expression of 5′-DI, which would lower T3 generation in liver. However, as noted above, activation of liver NFkB is not seen in NTIS.

Interleukin 6

Serum IL-6 is often elevated in NTIS,108 and its level is inversely related to T3 levels. Stouthard et al.109 gave recombinant human IL-6 chronically to human volunteers. Short-term infusion of IL-6 caused a suppression of TSH, but daily injections over 42 days caused only a modest decrease in T3 and a transient increase in rT3 and free T4 concentrations. IL-6 could be involved in the NTIS syndrome, although the mechanism was not defined. In an animal model of NTIS studied by Wiersinga and collaborators,110 antibody blockade of IL-6 failed to prevent the induced changes in thyroid hormone economy typical of NTIS. Boelen et al. studied the levels of IFN, IL-8, and IL-10 in patients with NTIS and found no evidence that they had a pathogenic role.111 Short-term administration of recombinant IFN-γ to normal subjects caused a minimal elevation of IL-6, no alteration in TNF, and did not significantly alter thyroid hormone levels. Michalaki et al. observed that serum T3 drops early after abdominal surgery as an early manifestation of the NTIS syndrome, prior to an increase in serum IL-6 or TNF-α, suggesting that these changes in cytokines do not induce the drop in T3.113

The potential interaction between cytokines and the hypothalamic-pituitary-thyroid axis is certainly complicated, and cytokines themselves operate in a network. For example, IL-1 and TNF can stimulate secretion of IL-6. Activation of TNF and IL-1 production is associated with the occurrence of cytokine inhibitors in serum, which are actually fragments of the cytokine receptor or actual receptor antagonists. Soluble TNF receptor and IL-1 RA are receptor antagonists, which can inhibit the function of the free cytokines. These molecules are increased in many infectious, inflammatory, and neoplastic conditions. Boelen et al.113 found evidence that the NTIS is “an acute phase response” generated by activation of a cytokine network. Soluble TNF, soluble TNF receptor, soluble IL-2 receptor antagonist, and IL-6 all inversely correlated with serum T3 levels.

While the studies noted fail to pinpoint one cytokine as the crucial mediator, we can be convinced that striking changes in cytokines co-occur during NTIS and probably play a pathogenic role by mechanisms yet undefined.

 

OTHER FACTORS ALTERING SERUM T4 SUPPLY

Altered CNS Metabolism

In healthy men going through two 4.5-hour-long sessions of induced hypoglycemia, TSH, fT3 and fT4 are significantly reduced.114 Perinatal asphyxia, recognized by low Apgar scores, is associated with a depression of TSH, T4, and T3, and the reductions are greatest in infants with hypoxic/ischemic encephalopathy. In this study, 6 of 11 infants with fT4 < 2ng/dL died. These data suggest, not surprisingly, that reduced substrate or O2 supply to the CNS could induce hypothalamic/pituitary dysfunction.114,115

Glucagon

Administration of glucagon to dogs caused a significant fall in serum T3, suggesting that stress-induced hyperglucagonemia may be a contributor to the NTIS syndrome by altering intracellular metabolism of T4.116

Dopamine

Dopamine given in support of renal function and cardiac function must play a role in many patients who develop low hormone levels while in an intensive care unit setting. Dopamine inhibits TSH secretion directly, depresses further the already abnormal thyroid hormone production, and induces significant worsening of the low hormone levels. Withdrawal of dopamine infusion is followed by a prompt dramatic elevation of TSH, a rise in T4 and T3, and an increase of the T3/rT3 ratio. All of these changes suggested to Van den Berghe et al.117 that dopamine makes some patients with NTIS hypothyroid, inducing a condition of iatrogenic hypothyroidism, and that treatment (presumably by administering thyroid hormone), “should be evaluated.”

Leptin

Leptin plays a key role in control of thyroid hormone levels during starvation in animals. During starvation, leptin levels drop. With this there is diminished stimulation of TRH, thus diminished secretion of TSH, and lowered thyroid hormone levels. Administration of leptin appears to work via the arcuate nucleus of the hypothalamus to induce production of pro-opiomelanocortin (POMC), and thus α-melanocyte-stimulating hormone (αMSH), and reduce Agouti-related protein (AgRP). Normally αMSH stimulates the melanocortin 4 receptor (MC4R), whereas AgRP suppresses it. Presumably through these actions, a lack of leptin during starvation leads to diminished stimulation of the MC4R receptor on the TRH neurons in ventricular nuclear centers and thus diminished TRH secretion. Administration of leptin partially reverses this sequence.118 These actions appear to be part of an energy-conserving scheme related to thyroid changes during starvation and are associated with leptin-induced increase in appetite, decreased energy expenditure, and modified neuroendocrine function. The relevance of this to human physiology is as yet unclear, but the data strongly suggest that leptin is involved in the down-regulation of thyroid function during acute starvation.118-120 In clinical trials, stimulation of growth hormone secretion by GH secretogogues lead to increased insulin and leptin levels in severely ill ICU patients. To date, studies of leptin levels in patients with NTIS have indicated they are normal or elevated, not low.121

Atrial Natriuretic Peptides

Atrial natriuretic peptides, including amino acids 1 to 30, amino acids 31 to 67 (known as vessel dilator), 79 to 98 (kaliuretic hormone), and 99 to 126 (atrial natriuretic hormone), derived from the ANH prohormone, significantly decreased circulating concentrations of total T4, free T4, and free T3, when given to healthy humans for 60 minutes. A reciprocal increase in TSH lasted for 2 or 3 hours after cessation of the administration of these hormones, suggesting that the effect was a direct inhibition of thyroid hormone release from the thyroid gland rather than an action of the hormones upon the hypothalamus or pituitary. No data are available on these factors in NTIS122 (Table 4).

 

DIAGNOSIS

Typically the endocrinologist is presented with a severely ill patient in whom there is no prior history suggestive of pituitary disease, in whom clinical findings of hypothyroidism are either absent or masked by other disorders, with a T4 and FTI (by an index method) that are low, a low or normal TSH, and, if measured, a low T3. If T4 is below 4 μg/dL in this setting, the diagnosis of NTIS, associated with a potentially fatal outcome, may be assumed; rT4 may be normal or elevated and is not diagnostic. An elevated TSH suggests the presence of prior hypothyroidism, which should be treated. Finding positive antithyroid antibody titers supports the diagnosis of primary hypothyroidism but does not prove it.

Serum cortisol should be measured. Transient, apparently central, hypoadrenalism may occur in severe illness.123-125 Cortisol should be above 20 μg/dL, and commonly is above 30. If below 20, ACTH should be drawn, and the patient may be given supportive cortisol therapy. Serum cortisol should certainly be determined if thyroid hormone is to be given. Since CBG may be reduced, it is advisable to measure serum free cortisol if possible. It is useful to determine FSH in postmenopausal women as a sign of pituitary function, but this is less clearly valuable in men. If there is a reason to consider hypopituitarism, a CAT scan of the pituitary is appropriate, or at least a skull film.

Use of aspirin, dilantin, and carbamazepine should be noted, since they can lower T4 and FTI as measured by several “index” methods. Dopamine used in the setting of severe illness can induce clear-cut hypothyroidism. Hyperthyroidism is the typical cause of suppression of TSH below 0.1 μU/mL, but it is rarely difficult to exclude this diagnosis in the setting of severely depressed T4 and T3.

 

IS THYROID HORMONE TREATMENT OF NTIS ADVANTAGEOUS OR DISADVANTAGEOUS?

Two valuable studies are available on replacement therapy using thyroid hormone in patients with NTIS. In the study by Brent and Hershman,54 replacement with 1.5 μg T4 IV per kilogram body weight daily, in 12 patients, promptly returned serum T4 levels to normal (thereby proving that a binding defect was not the cause of the low T4) but did not normalize T3 levels over a period of 2 to 3 weeks. However, in both the treated and control group, mortality was 80%.54 Clearly this excellent small study, which used for primary therapy what would now be considered the wrong hormone, failed to show either an advantageous or disadvantageous effect. It is possible that the failure to show a positive effect was due to the failure of T3 levels to be restored to normal. In a study of severely burned patients given 200 μg T3 daily, again there was no evidence of a beneficial or disadvantageous effect.126 Mortality was not so great, as in the Brent and Hershman study, but it is entirely possible that the high levels of T3 given worsened the hypermetabolism known to be present in burn patients and could have, at these levels, been disadvantageous.

An important study by Acker et al. certainly advises caution regarding T4 therapy in patients with acute renal failure. Numerous studies in animals have documented a beneficial effect of T4 therapy in experimental acute renal failure.127 In a randomized, controlled prospective study of patients with acute renal failure (ARF), treated patients received 150 μg of thyroxine a total of four times intravenously over 2 days.128 The single difference recognized in the subsequent laboratory data was a suppression of TSH. T4 treatment had no effect on any measure of ARF severity. Among other questions, it is not clear that serum T3 levels were ever altered. However, mortality was higher in the thyroxine group (43% versus 13%) than in the control group. It is of interest that, as the authors state, “the observed mortality in the controls in this study was less than that typically seen in our institution in ARF and ICU patients, whereas the 43% mortality noted in the thyroid group better approximates both our experience and that reported in the literature for ICU patients.” It will be difficult to replicate this study (although this reader believes it should be replicated). But it is uncertain whether the small dose of thyroxine administered over 2 days actually is related to the mortality, considering that the mortality in the treated group was that usually observed, whereas the control happened to have a much lower mortality.128 The same group has also studied the effect of thyroid hormone treatment on posttransplant acute tubular necrosis. T3 treatment during the posttransplant period did not alter outcome in a beneficial or derogatory manner.129

Studies from animals are often quoted in the literature as an argument against treatment of NTIS or for the therapy. A study of sepsis induced in animals showed no difference in mortality with treatment, but some animals treated with thyroid hormone died earlier than did those that were untreated.130 Chopra et al. induced NTIS in rats by injection of turpentine oil. The reduction in T4, T3, free T4 index, and TSH were associated with no clear evidence of tissue hypothyroidism, and urinary nitrogen excretion was normal. Thyroid hormone replacement with T4 or T3 did not significantly alter enzyme activities or urinary nitrogen excretion.131 Healthy pigs were subjected to 20 minutes of regional myocardial ischemia by Hsu and collaborators,132 and this was associated with a drop in T3, free T3, and elevated rT3. Some animals were treated with 0.2 μg T3 per kilogram for five doses over 2 hours. While myocardial infarction size was not altered, the pigs treated with T3 showed a more rapid improvement in cardiac index. Oxygen consumption did not alter. It should be noted that the T3 levels fell back to normal levels within 4 hours of the last T3 dose, suggesting that more prolonged therapy might have been beneficial. Katzeff et al.133 studied a model of NTIS induced by caloric restriction in young rats. In these animals, T3 was reduced, and there was a decrease in LV relaxation time, SERCA2 mRNA, and αMHC mRNA. All changes were reversed to normal values by supplementation with T3, suggesting that the low-T3 syndrome was related to the pathologic cardiac changes. Sepsis and multisystem organ failure are often associated with disseminated intravascular coagulation and consumption of coag inhibitors such as antithrombin III. Chapital studied a model of sepsis in rats and showed that T3 supplementation reduced the decrease in antithrombin III levels, which presumably would reflect a beneficial effect.134 Dogs subjected to hemorrhagic shock recovered more cardiovascular function when given T3 intravenously than did untreated animals.135 Neurologic outcome after anoxia is improved in dogs by T3 treatment.136

Short-term studies on T3 replacement of patients in shock, in patients with respiratory disease, in subjects who are brain dead and potential organ donors, and in patients undergoing coronary artery bypass grafts all suggest modest cardiovascular benefits from the administration of T3. One study reports benefit by replacing T3 to elevate the depressed T3 levels in premature infants.137 Other studies found no apparent effects. Children treated with T3 postoperatively when they have undergone cardiac surgery also require less cardiac support(138). T3 administration (one dose of approximately 6 μg IV) did not alter cardiac performance in brain-dead transplant donors.139 Coronary artery bypass, as studied by Klemperer and collaborators,36 was associated with a drop in serum T3; IV administration of T3 elevated T3 above normal, augmented cardiac output, and reduced the need for pressor support but had no other effect. In this study, however, the patients had a very favorable prognosis and minimal NTIS, so the study primarily shows that administration of T3 had no adverse effect under these circumstances. In a study of patients after heart transplant, patients with the low T3 syndrome (NTIS) had higher mortality, higher incidence of acute rejection, highest number of re-operations, and higher incidence of infections, compared to those without NTIS (140). In a study reported several years ago, T3 administration to critically ill neonates with severe respiratory distress appeared to improve survival. Infants of less than 37 weeks gestational age or weighing less than 220 grams were given prophylactic doses of thyroxine and T3 daily and had a lower mortality rate than untreated infants.137 Use of thyroid hormone replacement in children after cardiac surgery has been extensively reviewed by Haas et al., with the conclusion that it is a desirable treatment option, especially in high-risk patients.141 Goarin et al. studied the effect of T3 administration in brain-dead organ donors and found that although it returned T3 levels to normal, it did not improve hemodynamic status or myocardial function.142 Pingitore et al. gave T3 by IV infusion for 3 days to patients with chronic heart failure. Heart rate, plasma nor-epinephrine (down 52%), natriuretic peptide, and aldosterone (down 23%) were all significantly diminished, and ventricular performance improved, without side effects.143 In a randomized study of patients for 24 hours after coronary bypass, correction of the usual drop in serum T3 by IV T3 infusion had no beneficial or deleterious effect on cardiac parameters.144 Of interest, it also did not affect leucine flux or urinary nitrogen excretion, contrary to the usual assumption that a drop in serum T3 should spare body protein. Novitsky (145) studied three conditions in which NTIS and myocardial functional depression have been documented - i) transient regional myocardial ischemia and reperfusion, ii) transient global myocardial ischemia in patients undergoing cardiac surgery on cardiopulmonary bypass, and iii) transient inadequate global myocardial perfusion in brain-dead potential organ donors. Under all three conditions, in models and in man, following administration of T3/T4, the myocardial dysfunction was rapidly reversed. Cautiously use of thyroid hormonal therapy to any patient with the ESS and/or a stunned myocardium was advocated. The general outcome of these studies is that they weakly support the use of T3, and none of the studies found evidence of damage caused by treatment.144-150

In summary, it can be stated that there is no clear evidence that thyroxine or triiodothyronine treatment of NTIS in animals or man is disadvantageous, but no certain proof that it is advantageous.  In the acute NTIS syndrome associated with operations, short term treatment with T3 augments cardiac function, but has not been shown to alter the already low mortality (150). However, what evidence there is suggests TH may be beneficial. The argument has been raised that administration of thyroid hormone in NTIS would prevent the elevation in TSH commonly seen in recovering patients. This seems rather specious. More objectively, the elevation of TSH is another suggestion that the few patients who survive the ordeal were hypothyroid and left untreated. Lastly, it is unlikely that administration of replacement hormone during NTIS would be harmful, even if all of the evidence presented suggesting hypothyroidism was erroneous, and the patients were in fact euthyroid.

 

IF THYROID HORMONE REPLACEMENT IS GIVEN, WHAT SHOULD IT BE?

Clearly the high mortality rate in patients in the chronic phase of NTIS, with T4 under 4 μg/dL, suggests that this is a target group in whom thyroid hormone administration should be considered. In this group of patients, there appears to be no obvious contraindication to replacement therapy, with the possible exception of people who have cardiac decompensation or arrhythmias. Even here, the evidence is uncertain. There is no clear evidence that administration of replacement doses of T3 to patients with low cardiac output is disadvantageous, and in fact current studies using intravenous T3 in these patients indicate it is well tolerated and may be beneficial.151 Arrhythmias obviously also raise a question, but again, there is no evidence that replacement of thyroid hormone to a normal level would cause trouble in control of arrhythmias. Low free T3 levels are reported to be associated with an increased incidence of fibrillation after cardiac surgery in elderly patients.152 Thus even in this group of patients, it is reasonable to suggest therapy. It should also be noted that among patients with NTIS, there will certainly be patients who are clearly hypothyroid—based on known disease, treatment with dopamine, or elevated TSH—who need replacement therapy by any standard.

If therapy is to be given, it cannot be thyroxine alone, since this would fail to promptly elevate T3 levels.54 Treatment should include oral, or if this is impractical, intravenous T3, and probably should be at the replacement level of approximately 50 μg/day given in divided doses. It may be appropriate to give slightly higher doses, such as 75 μg/day for 3 to 4 days to increase the body pool more rapidly, followed by replacement doses as described. Coincidentally, it is appropriate to start replacement with T4. Serum levels of T4 and T3 should be followed at frequent intervals (every 48 hours) and dosages adjusted to achieve a serum T3 level at least low normal (70 to 100 ng/dL) prior to the next scheduled dose. If treatment is successful, T3 administration can gradually be reduced, and thyroxine administration can be increased to replacement levels as deiodination increases. Because of the marked diminution in T4 to T3 deiodination, and shunting of T4 toward r T3, replacement with T4 may initially only lead to elevation of rT3 and have very little effect upon T3 levels, or physiologic action. In this situation, continued administration of T3 would be preferred. An alternate therapy, giving TRH to stimulate TSH production and TH release, has been shown to be effective in raising TH levels during short term treatment. This is described below. No prolonged treatment, or effects on survival have so far been reported.

 

ADDITIONAL SUPPORTIVE HORMONAL THERAPY TO CONSIDER

Although this discussion concentrates on the potential value of treating patients with NTIS with replacement thyroid hormone, several important recent studies expand the concept to other areas, including treatment of the associated hyperglycemia, relative adrenal insufficiency, use of beta blockers in burn patients, and possible use of GHRH and testosterone. Van den Berghe and co-workers have suggested that the acute and prolonged critical illness responses are entirely different neuroendocrine conditions. In protracted severe illness, patients are kept alive with conditions that previously caused death. However, this process has unmasked a variety of nonspecific wasting syndromes that include protein loss, accumulation of fat stores, hyperglycemia and insulin resistance, hypoproteinemia, hypercalcemia, potassium depletion, and hypertriglyceridemia. In prolonged illness, cortisol values are elevated, although ACTH levels are low, indicating that other mechanisms are driving the steroid response. Growth hormone secretory pulses are reduced, and the mean GH concentration is low in prolonged critical illness. FSH and LH are reduced, and testosterone levels are reduced. These authors maintain that the reduced neuroendocrine drive, present in the chronic phase of illness in an intensive care setting, is unlikely to be an evolutionary preserved beneficial process. They suggest that the administration of hypothalamic physiotropic releasing peptides may be a safer strategy than the administration of peripherally active hormones.153-156

NTIS is typically associated with poor nutrition unless supportive measures are taken, and undernutrition is a known inducer of NTIS. Yet nutritional support is not uncomplicated. Parenteral feeding of rabbits (in contrast to fasting) in a model of chronic NTIS resulted in a normalization of low T3 levels, but did not correct low T4 levels(157) .Use of early parenteral alimentation for patients in the ICU with NTIS was recommended by Perez-Guisado et al (158), who found it decreased length of hospital stay.  However a study by Langouche et al (159) found, in contrast, that adding parenteral nutrition sooner than one week in the ICU increased complications and delayed recovery.

  Hyperglycemia and insulin resistance are common in critically ill patients, even if they have not previously had diabetes. Van den Berghe et al carried out a prospective randomized study on ICU patients on mechanical ventilation, maintaining blood glucose at a level between 80 and 110 mg/dl, versus allowing glucose to range between a level of 180 – 200 mg/dl. Intensive insulin therapy reduced overall in-hospital mortality by 34 percent, bloodstream infections by 46 percent, acute renal failure requiring dialysis or hemofiltration by 41 percent, the median number of red-cell transfusions by 50 percent, and critical-illness polyneuropathy by 44 percent, and patients receiving intensive therapy were less likely to require prolonged mechanical ventilation and intensive care (98). This strict blood sugar control can lead to bouts of hypoglycemia, and higher controlled values are currently aimed for. In isolated brain injury patients, intensive insulin therapy reduced mean and maximal intracranial pressure while identical cerebral perfusion pressures were obtained with eightfold less vasopressors. Seizures and diabetes insipidus occurred less frequently. At 12 months follow-up, more brain-injured survivors in the intensive insulin group were able to care for most of their own needs. Preventing even moderate hyperglycemia with insulin during intensive care protected the central and peripheral nervous systems, with clinical consequences such as shortening of intensive care dependency and possibly better long-term rehabilitation. Prevention of catabolism, acidosis, excessive inflammation, and impaired innate immune function may explain previously documented beneficial effects of intensive insulin therapy on outcome of critical illness.   Severe burns are known to be associated with a hypermetabolic state and a strong sympathetic response. Beta blockade given as propranolol to reduce the resting heart rate by 20% decreased resting energy expenditure and increased net muscle protein balance  significantly in a group of burn patients. It is logical that this would be a significant benefit (160). Severe sepsis, which is of course associated with NTIS, is frequently associated with relative adrenal insufficiency, and possibly systemic inflammation-induced glucocorticoid receptor resistance. In a prospective randomized study, Annane et al studied a seven day treatment of patients with septic shock, by giving hydrocortisone, 50 mg q6h, and 9-alpha-fludrocortisone, 50 mg once daily. The risk of death in this treated group was significantly reduced without increasing any adverse effects. The treatment was most beneficial in individuals who responded poorly to a 250 mg ACTH test, which was conducted prior to the therapy.  Non-response was defined as a response of 9 mg/dl or less, between the lowest, and highest concentration taken after the ACTH injection. Samples were taken in this study at 30 and 60 minutes (161). The severity of the illness was suggested by the statistics that 63% died in the placebo group, and 53% in the corticosteroid treatment group. The authors recommend that all patients with catecholamine dependent septic shock should be given a combination of hydrocortisone and fludrocortisone as soon as a short corticotrophin stimulation test is performed, and the treatment should be continued for seven days in non-responders. Hamrahian et al advise caution in using total serum cortisol measurements in patients with serum albumin levels below 21.5gm/dl. They observed that these patients may have low total cortisol because of low CBG, but have normal or elevated free cortisol levels (162) In contrast to the generally beneficial effects of hormonal therapy described above, high levels of growth hormone given to critically ill patients were found by Takala et al to augment mortality. The dosage used was 0.1 mg/kg bw, for up to 21 days. Mortality rate was nearly double. These authors suggest that GH may have an adverse effect upon immunity, cause fluid retention, and cause hyperglycemia (163).

TREATMENT WITH HYPOTHALAMIC RELEASING HORMONES

Van Den Berghe and collaborators have pioneered studies on the effects of hypothalamic releasing hormones in patients with severe NTIS. The logic supporting this approach is that it corrects a major cause of the low hormonal state, and may allow normal feed-back control and peripheral regulation of hormones, thus being more physiological than replacing the peripheral hormone deficit directly. Extensive studies document restoration of T4 and T3 levels following administration of TRH and GH secretagaugue (153). In a rabbit model of NTIS treatment with GHRP-2 and TRH reactivated the GH and TSH axes and altered liver deiodinase activity, driving T4 to T3 conversion. In NTIS there are suppressed pulsatile GH, TSH, LH secretion in the face of low serum concentrations of IGF-I, IGFBP-3 and the acid-labile subunit (ALS), thyroid hormones, and total and estimated free testosterone levels, whereas free estradiol (E2) estimates are normal. Ureagenesis and breakdown of bone tissue are increased. Baseline serum TNF-alpha, IL-6 and C-reactive protein level and white blood cell (WBC) count are elevated; serum lactate is normal. Coadministration of GHRP-2, TRH and GnRH reactivated the GH, TSH and LH axes in prolonged critically ill men and evoked beneficial metabolic effects which were absent with GHRP-2 infusion alone and only partially present with GHRP-2 + TRH. These data underline the importance of correcting the multiple hormonal deficits in patients with prolonged critical illness to counteract the hypercatabolic state (154. Contrary to expectation, intensive insulin therapy suppressed serum IGF-I, IGFBP-3, and acid-labile subunit concentrations. This effect was independent of survival of the critically ill patient. Concomitantly, serum GH levels were increased by intensive insulin therapy. The data suggest that intensive insulin therapy surprisingly suppressed the somatotropic axis despite its beneficial effects on patient outcome. GH resistance accompanied this suppression of the IGF-I axis. To what extent and through which mechanisms the changes in the GH-IGF-IGFBP axis contributed to the survival benefit under intensive insulin therapy remain elusive (155). While outcome studies using this approach are not available, it is quite possible that treatment of NTIS by use of hypothalamic releasing hormones may be a preferred approach.

CONCLUSIONS

This review has presented the arguments for administration of replacement T3 and T4 hormone in patients with NTIS. However, it is impossible to be certain at this time that it is beneficial to replace hormone, or whether this could be harmful. Other recent reviews on this topic are available (164). Only a prospective study will be adequate to prove or disprove the value of hormone replacement, and probably this would need to involve hundreds of patients. Tragically, many ICU patients continuing to die with NTIS (we do not know if this is from NTIS) and we have now waited over 40 years for the proper controlled study to be done. One cannot envisage that replacement of thyroxin or T3 can “cure” patients with NTIS. The probable effect, if any is achieved, will be a modest increment in overall physiologic function and a decrease in mortality. Perhaps this would be 5%, 10%, or 20%. If effective, thyroid hormone replacement will be one of many beneficial treatments given the patient, rather than a single magic bullet which would reverse all the metabolic changes going wrong in these severely ill patients. Ongoing studies document the beneficial effects of hormone replacement in these acutely and severely ill patients. Possibly therapy will ultimately involve replacement of peripheral hormones, or may instead be via growth hormone-releasing peptide (GHRP), TRH, GnRH, insulin, adrenal steroids, and leptin.

  REFERENCES

1.         McIver B, Gorman CA: Euthyroid sick syndrome: An overview, Thyroid 7:125–132, 1997.

2.         DeGroot LJ: “Non-thyroidal illness syndrome” is functional central hypothyroidism, and if severe, hormone replacement is appropriate in light of present knowledge, J Endocrinol Invest 26:1163–1170, 2003.

3.         Stathatos N, Wartofsky L: The euthyroid sick syndrome: is there a physiologic rationale for thyroid hormone treatment? J Endocrinol Invest 26:1174–1179, 2003.

4.         Hennemann G, Docter R, Krenning EP: Causes and effects of the low T3 syndrome during caloric deprivation and non-thyroidal illness: an overview, Acta Med Kaust 15:42–45, 1988.

5.         Phillips RH, Valente WA, Caplan ES, et al: Circulating thyroid hormone changes in acute trauma: prognostic implications for clinical outcome, J Trauma 24:116–119, 1984.

6.         Vardarli I, Schmidt R, Wdowinski JM, et al: The hypothalamo-hypophyseal-thyroid axis, plasma protein concentrations and the hypophyseogonadal axis in low T3 syndrome following acute myocardial infarct, Klin Wochenschrift 65:129–133, 1987.

7.         Eber B, Schumacher M, Langsteger W, et al: Changes in thyroid hormone parameters after acute myocardial infarction, Cardiology 86:152–156, 1995.

8.         Holland FW, Brown PS, Weintraub BD, et al: Cardiopulmonary bypass and thyroid function: a “euthyroid sick syndrome.” Ann Thorac Surg 52:46–50, 1991.

9.         Vexiau P, Perez-Castiglioni P, Socie G, et al: The “euthyroid sick syndrome”: Incidence, risk factors and prognostic value soon after allogeneic bone marrow transplantation, Br J Hematol 85:778–782, 1993.

10.       Harris ARC, Fang SL, Vagenakis AG, et al: Effect of starvation, nutriment replacement, and hypothyroidism on in vitro hepatic T4 to T3 conversion in the rat, Metabolism 27:1680–1690, 1978.

11.       Welle SL, Campbell RG: Decrease in resting metabolic rate during rapid weight loss is reversed by low-dose thyroid hormone treatment, Metabolism 35:289–291, 1986.

12.       Plikat K, Langgartner J, Buettner R, et al: Frequency and outcome of patients with nonthyroidal illness syndrome in a medical intensive care unit, Metabolism 56(2):239–244, 2007 Feb.

13.       Girvent M, Maestro S, Hernandez R, et al: Euthyroid sick syndrome, associated endocrine abnormalities, and outcome in elderly patients undergoing emergency operation, Surgery 123:560–567, 1998.

14.       Wartofsky L, Burman KD: Alterations in thyroid function in patients with systemic illnesses: the “Euthyroid Sick Syndrome”, Endocrine Rev 3:164–217, 1982.

15.       Kaptein EM: Clinical relevance of thyroid hormone alterations in nonthyroidal illness, Thyroid International 4:22–25, 1997.

16.       Docter R, Krenning EP, de Jong M, et al: The sick euthyroid syndrome: changes in thyroid hormone serum parameters and hormone metabolism, Clin Endocrinol 39:499–518, 1993.

17.       Chopra IJ, Huang TS, Boado R, et al: Evidence against benefit from replacement doses of thyroid hormones in nonthyroidal illness: studies using turpentine oil–injected rat, J Endocrinol Invest 10:559–564, 1987.

18.       Schilling JU, Zimmermann T, Albrecht S, et al: Low T3 syndrome in multiple trauma patients – a phenomenon or important pathogenetic factor? Medizinische Klinik 3:66–69, 1999.

19.       Schulte C, Reinhardt W, Beelen D, et al: Low T3-syndrome and nutritional status as prognostic factors in patients undergoing bone marrow transplantation, Bone Marrow Transplantation 22:1171–1178, 1998.

20.       Girvent M, Maestro S, Hernandez R, et al: Euthyroid sick syndrome, associated endocrine abnormalities, and outcome in elderly patients undergoing emergency operation, Surgery 123:560–567, 1998.

21.       Maldonado LS, Murata GH, Hershman JM, et al: Do thyroid function tests independently predict survival in the critically ill? Thyroid 2:119, 1992.

22.       Vaughan GM, Mason AD, McManus WF, et al: Alterations of mental status and thyroid hormones after thermal injury, J Clin Endocrinol Metab 60:1221, 1985.

23.       De Marinis L, Mancini A, Masala R, et al: Evaluation of pituitary-thyroid axis response to acute myocardial infarction, J Endocrinol Invest 8:507, 1985.

24.       Surks MI, Hupart KH, Pan C, et al: Normal free thyroxine in critical nonthyroidal illnesses measured by ultrafiltration of undiluted serum and equilibrium dialysis, J Clin Endocrinol Metab 67:1031–1039, 1988.

25.       Melmed S, Geola FL, Reed AW, et al: A comparison of methods for assessing thyroid function in nonthyroidal illness, J Clin Endocrinol Metab 54:300–306, 1982.

26.       Kaptein EM, MacIntyre SS, Weiner JM, et al: Free thyroxine estimates in nonthyroidal illness: comparison of eight methods, J Clin Endocrinol Metab 52:1073–1077, 1981.

27.       Kantor M, et al: Admission thyroid evaluation in very-low-birth-weight infants: association with death and severe intraventricular hemorrhage, Thyroid 13:965, 2003.

28.       Chopra IJ, Solomon DH, Hepner GW, et al: Misleadingly low free thyroxine index and usefulness of reverse triiodothyronine measurement in nonthyroidal illnesses, Ann Int Med 90:905–912, 1979.

29.       Bacci V, Schussler GC, Kaplan TB: The relationship between serum triiodothyronine and thyrotropin during systemic illness, J Clin Endocrinol Metab 54:1229–1235, 1982.

30.       Sapin R, Schlienger JL, Kaltenbach G, et al: Determination of free triiodothyronine by six different methods in patients with nonthyroidal illness and in patients treated with amiodarone, Ann Clin Biochem 32:314–324, 1995.

31.       Chopra IJ, Taing P, Mikus L: Direct determination of free triiodothyronine (T3) in undiluted serum by equilibrium dialysis/radioimmunoassay, Thyroid 6:255–259, 1996.

32.       Chopra IJ: Simultaneous measurement of free thyroxine and free 3,5,3’-triiodothyronine in undiluted serum by direct equilibrium dialysis/radioimmunoassay: evidence that free triiodothyronine and free thyroxine are normal in many patients with the low triiodothyronine syndrome, 1998.

33.       Faber J, Kirkegaard C, Rasmussen B, et al: Pituitary-thyroid axis in critical illness, J Clin Endocrinol Metab 65(2):315–320, 1987 Aug.

34.       Fritz KS, Wilcox RB, Nelson JC: Quantifying spurious free T4 results attributable to thyroxine-binding proteins in serum dialysates and ultrafiltrates, Clin Chem 53(5):985–988, 2007 May.

35.       Peeters RP, Wouters PJ, Kaptein E, et al: Reduced activation and increased inactivation of thyroid hormone in tissues of critically ill patients, J Clin Endocrinol Metab 88(7):3202–3211, 2003 Jul.

36.       Klemperer JD, Klein I, Gomez M, et al: Thyroid hormone treatment after coronary-artery bypass surgery, N Engl J Med 333:1522–1527, 1995.

37.       Osburne RC, Myers EA, Rodbard D, et al: Adaptation to hypocaloric feeding: physiologic significance of the fall in T3, Metabolism 32:9–13, 1983.

38.       Afandi B, Schussler GC, Arafeh A-H, et al: Selective consumption of thyroxine-binding globulin during cardiac bypass surgery, Metabolism 49:270–274, 2000.

39.       Jirasakuldech B, Schussler GC, Yap MG, et al: A characteristic serpin cleavage product of thyroxine-binding globulin appears in sepsis sera, J Clin Endocrinol Metab 85:3996–3999, 2000.

40.       den Brinker M, Joosten KF, Visser TJ, et al: Euthyroid sick syndrome in meningococcal sepsis: the impact of peripheral thyroid hormone metabolism and binding proteins, J Clin Endocrinol Metab 90(10):5613–5620, 2005.

41.       Uchimura H, Nagataki S, Tabuchi T, et al: Measurements of free thyroxine: comparison of percent of free thyroxine in diluted and undiluted sera, J Clin Endocrinol Metab 42:561–566, 1976.

42.       Nelson JC, Weiss RM: The effect of serum dilution on free thyroxine concentration in the low T4 syndrome of nonthyroidal illness, J Clin Endocrinol Metab 61:239–246, 1985.

43.       Wang R, Nelson JC, Weiss RM, et al: Accuracy of free thyroxine measurements across natural ranges of thyroxine binding to serum proteins, Thyroid 10:31–39, 2000.

44.       Kaptein EM: Thyroid hormone metabolism and thyroid diseases in chronic renal failure, Endocrine Rev 17:45–63, 1996.

45.       Liewendahl K, Helenius T, Naveri H, et al: Fatty acid-induced increase in serum dialyzable free thyroxine after physical exercise: implication for nonthyroidal illness, J Clin Endocrinol Metab 74:1361–1365, 1992.

46.       Mendel CM, Frost PH, Cavalieri RR: Effect of free fatty acids on the concentration of free thyroxine in human serum: the role of albumin, J Clin Endocrinol Metab 63:1394–1399, 1986.

47.       Jaume JC, Mendel CM, Frost PH, et al: Extremely low doses of heparin release lipase activity into the plasma and can thereby cause artifactual elevations in the

48.       Wang Y-S, Hershman JM, Pekary AE: Improved ultrafiltration method for simultaneous measurement of free thyroxine and free triiodothyronine in serum, Clin Chem 31:517–522, 1985.

49.       Mendel CM, Laufhton CW, McMahon FA, et al: Inability to detect an inhibitor of thyroxine-serum protein binding in sera from patients with nonthyroidal illness, Metabolism 40:491–502, 1991.

50.       Chopra IJ, Huang T-S, Beredo A, et al: Evidence for an inhibitor of extrathyroidal conversion of thyroxine to 3,5,3’-triiodothyronine in sera of patients with nonthyroidal illnesses, J Clin Endocrinol Metab 60:666, 1985.

51.       Wang R, Nelson JC, Wilcox RB: Salsalate administration—a potential pharmacological model of the sick euthyroid syndrome, J Clin Endocrinol Metab 83:3095–3099, 1998.

52.       Csako G, Zweig MH, Benson C, et al: On the albumin-dependence of the measurement of free thyroxine. II. Patients with nonthyroidal illness, Clin Chem 33:87–92, 1987.

53.       Chopra IJ, Chua Teco GN, Mead JF, et al: Relationship between serum free fatty acids and thyroid hormone binding inhibitor in nonthyroid illnesses, J Clin Endocrinol Metab 60:980–984, 1985.

54.       Brent GA, Hershman JM: Thyroxine therapy in patients with severe nonthyroidal illnesses and lower serum thyroxine concentration, J Clin Endocrinol Metab 63:1–8, 1986.

55.       Chopra IJ: Nonthyroidal illness syndrome or euthyroid sick syndrome? Endocrine Pract 2:45–52, 1996.

56.       Franklyn JA, Black EG, Betteridge J, et al: Comparison of second and third generation methods for measurement of serum thyrotropin in patients with overt hyperthyroidism, patients receiving thyroxine therapy, and those with nonthyroidal illness, J Clin Endocrinol Metab 78:1368–1371, 1994.

57.       Vierhapper H, Laggner A, Waldhausl W, et al: Impaired secretion of TSH in critically ill patients with “low T4-syndrome”. Acta Endocrinol 101:542–549, 1982.

58.       Faber J, Kirkegaard C, Rasmussen B, et al: Pituitary-thyroid axis in critical illness, J Clin Endocrinol Metab 65:315–320, 1987.

59.       Arem R, Deppe S: Fatal nonthyroidal illness may impair nocturnal thyrotropin levels, Am J Med 88:258–262, 1990.

60.       Lee H-Y, Suhl J, Pekary AE, et al: Secretion of thyrotropin with reduced concanavalin-A-binding activity in patients with severe nonthyroid illness, J Clin Endocrinol Metab 65:942, 1987.

61.       Spratt DI, Bigos ST, Beitins I, et al: Both hyper- and hypogonadotropic hypogonadism occur transiently in acute illness: bio- and immunoactive gonadotropins, J Clin Endocrinol Metab 75:1562–1570, 1992.

62.       Spratt DI, Cox P, Orav J, et al: Reproductive axis suppression in acute illness is related to disease severity, J Clin Endocrinol Metab 76:1548–1554, 1993.

63.       Van den Berghe G, Weekers F, Baxter RC, et al: Five-day pulsatile gonadotropin-releasing hormone administration unveils combined hypothalamic-pituitary-gonadal defects underlying profound hypoandrogenism in men with prolonged critical illness, J Clin Endocrinol Metab 86:3217–3226, 2001.

64.       Kaptein EM, Grieb DA, Spencer CA, et al: Thyroxine metabolism in the low thyroxine state of critical non-thyroidal illnesses, J Clin Endocrinol Metab 53:764–771, 1981.

65.       Kaptein EM, Robinson WJ, Grieb DA, et al: Peripheral serum thyroxine, triiodothyronine and reverse triiodothyronine kinetics in the low thyroxine state of acute nonthyroidal illnesses. A noncompartmental analysis, J Clin Invest 69:526–535, 1982.

66.       Lim VS, Fang VS, Katz AI, et al: Thyroid dysfunction in chronic renal failure. A study of the pituitary-thyroid axis and peripheral turnover, Kinetics of thyroxine and triiodothyronine, J Clin Invest 60(3):522–534, 1997.

67.       Lim C-F, Docter R, Visser TJ, et al: Inhibition of thyroxine transport into cultured rat hepatocytes by serum of nonuremic critically ill patients: effects of bilirubin and nonesterified fatty acids, J Clin Endocrinol Metab 76:1165–1172, 1993.

68.       Vos RA, de Jong M, Bernard BF, et al: Impaired thyroxine and 3,5,3’-triiodothyronine handling by rat hepatocytes in the presence of serum of patients with nonthyroidal illness, J Clin Endocrinol Metab 80:2364–2370, 1995.

69.       Lim C-F, Docter R, Krenning EP, et al: Transport of thyroxine into cultured hepatocytes: effects of mild nonthyroidal illness and calorie restriction in obese subjects, Clinical Endocrinol 40:79–85, 1994.

70.       Sarne DH, Refetoff S: Measurement of thyroxine uptake from serum by cultured human hepatocytes as an index of thyroid status: reduced thyroxine uptake from serum of patients with nonthyroidal illness, J Clin Endocrinol Metab 61:1046–1052, 1985.

71.       Krenning D, et al: Decreased transport of thyroxine (T4), 3, 33′, 5-triiodothyronine (T3) and 3,33′,5′-triiodothyronine (rT3) into rat hepatocytes in primary culture due to a decrease of cellular ATP content and various drugs, FEBS Lett 140:229–233, 1982.

72.       Arem R, Wiener GJ, Kaplan SG, et al: Reduced tissue thyroid hormone levels in fatal illness, Metabolism 42:1102–1108, 1993.

73.       Peeters RP, van der Geyten S, Wouters PJ, et al: Tissue thyroid hormone levels in critical illness, J Clin Endocrinol Metab 90(12):6498–6507. Epub 2005 Sep 20, 2005.

74.       d’Amati G, di Gioia CRT, Mentuccia D, et al: Increased expression of thyroid hormone receptor isoforms in end-stage human congestive heart failure, J Clin Endocrinol Metab 86:2080–2084, 2001.

75.       Rodriguez-Perez A, Palos-Paz F, Kaptein E, et al: Identification of molecular mechanisms related to nonthyroidal illness syndrome in skeletal muscle and adipose tissue from patients with septic shock, Clin Endocrinol (Oxf) 68(5):821–827, 2008.

76.       Sanchez B, Jolin T: Triiodothyronine-receptor complex in rat brain: effects of thyroidectomy, fasting, food restriction, and diabetes, Endocrinology 129:361–367, 1991.

77.       Beigneux A, et al: Sick euthyroid syndrome is associated with decreased TR expression and DNA binding in mouse liver, Am J Physiol Endocrinol Metab 284:E228-E236, 2003.

78        -Lado-Abeal J, Romero A, Castro-Piedras I, Rodriguez-Perez A, Alvarez-Escudero J     Thyroid hormone receptors are down-regulated in skeletal muscle of patients with non-thyroidal illness syndrome secondary to non-septic shock. Eur J Endocrinol. 2010 Nov;163(5):765-73. 79        -Boelen A, Kwakkel J, Fliers E  Beyond low plasma T3: local thyroid hormone metabolism during inflammation and infection.Endocr Rev. 2011 Oct;32(5):670-93.

80.       .Mebis L, Debaveye Y, Ellger B, Derde S, Ververs EJ, Langouche L, Darras VM, Fliers E, Visser TJ, Van den Berghe G. Changes in the central component of the hypothalamus-pituitary-thyroid axis in a rabbit model of prolonged critical illness.Crit Care. 2009;13(5):R147

81.       Mebis L, Paletta D, Debaveye Y, Ellger B, Langouche L, D'Hoore A, Darras VM, Visser TJ, Van den Berghe G. Expression of thyroid hormone transporters during critical illness. Eur J Endocrinol. 2009 Aug;161(2):243-50.

82.       Debaveye Y, Ellger B, Mebis L, Visser TJ, Darras VM, Van den Berghe G.--Effects of substitution and high-dose thyroid hormone therapy on deiodination, sulfoconjugation, and tissue thyroid hormone levels in prolonged critically ill rabbits.Endocrinology. 2008 Aug;149(8):4218-28. .

83.       Castro I, Quisenberry L, Calvo RM, Obregon MJ, Lado-Abeal J. Septic shock non-thyroidal illness syndrome causes hypothyroidism and conditions for reduced sensitivity to thyroid hormone. J Mol Endocrinol. 2013 Mar 18;50(2):255-66.

84.       Brent GA, Hershman JM, Reed AW, et al: Serum angiotensin converting enzyme in severe nonthyroidal illness associated with low serum thyroxine concentration, Ann Intern Med 100:680–683, 1986.

84.       Seppel T, Becker A, Lippert F, et al: Serum sex hormone-binding globulin and osteocalcin in systemic nonthyroidal illness associated with low thyroid hormone concentrations, J Clin Endocrinol Metab 81:1663–1665, 1996.

86.       Chapital AD, Hendrick SR, Lloyd L, et al: The effects of triiodothyronine augmentation on antithrombin III levels in sepsis, Am Surg 67(3):253–255, 2001 Mar.

87.       Kaplan MM: Subcellular alterations causing reduced hepatic thyroxine-53-monodeiodinase activity in fasted rats, Endocrinology 104:58–64, 1979.

88.       Berger MM, Reymond MJ, Shenkin A, et al: Influence of selenium supplements on the post-traumatic alterations of the thyroid axis: a placebo-controlled trial, Intensive Care Med 27:91–100, 2001.

89.       Blake NG, Eckland JA, Foster OJF, et al: Inhibition of hypothalamic thyrotropin-releasing hormone messenger ribonucleic acid during food deprivation, Endocrinology 129:2714–2718, 1991.

90.       Fliers E, Guldenaar SEF, Wiersinga WM, et al: Decreased hypothalamic thyrotropin-releasing hormone gene expression in patients with nonthyroidal illness, J Clin Endocrinol Metab 82:4032–4036, 1997.

91.       Van den Berghe G, De Zegher F, Baxter RC, et al: Neuroendocrinology of prolonged critical illness: effects of exogenous thyrotropin-releasing hormone and its combination with growth hormone secretagogues, J Clin Endocrinol Metab 83:309–319, 1998.

92.       Nicoloff JT, Fisher DA, Appleman MD Jr: The role of glucocorticoids in the regulation of thyroid function in man, J Clin Invest 49:1922, 1970.

93.       Brabant G, Brabant A, Ranft U: Circadian and pulsatile thyrotropin secretion in euthyroid man under the influence of thyroid hormone and glucocorticoid administration, J Clin Endocrinol Metab 65:83, 1987.

94.       Benker G, Raida M, Olbricht T, et al: TSH secretion in Cushing’s syndrome: relation to glucocorticoid excess, diabetes, goiter, and the “sick euthyroid syndrome”, Clin Endocrinol 33:777–786, 1990.

95.       Bianco AC, Nunes MT, Hell NS, et al: The role of glucocorticoids in the stress-induced reduction of extrathyroidal 3,5,3′-triiodothyronine generation in rats, Endocrinology 120:1033–1038, 1987.

96.       Lim VS, Passo C, Murata Y, et al: Reduced triiodothyronine content in liver but not pituitary of the uremic rat model: demonstration of changes compatible with thyroid hormone deficiency in liver only, Endocrinology 114:280–286, 1984.

97.       Van den Berghe G, de Zegher F, Bouillon R: Acute and prolonged critical illness as different neuroendocrine paradigms, J Clin Endocrinol Metab 83:1827–1834, 1998.

94.       Van den Berghe G, et al: Intensive insulin therapy in critically ill patients, N Engl J Med 345:1359–1367, 2001.

98.       Van den Berghe G, Schoonheydt K, Becx P, et al: Insulin therapy protects the central and peripheral nervous system of intensive care patients, Neurology 64(8):1348–1353, 2005 Apr 26.

99.       Mönig H, Arendt T, Meyer M, et al: Activation of the hypothalamo-pituitary-adrenal axis in response to septic or non-septic diseases—implications for the euthyroid sick syndrome, Intensive Care Med 25:1402–1406, 1999.

100.     Hermus RM, Sweep CG, van der Meer MJ, et al: Continuous infusion of interleukin-1 beta induces a non-thyroidal illness syndrome in the rat, Endocrinology 131:2139–2146, 1992.

101.     Cannon JG, Tompkins RG, Gelfand JA, et al: Circulating interleukin-1 and tumor necrosis factor in septic shock and experimental endotoxin fever, J Infect Dis 161:79–84, 1990.

102.     van der Poll T, Van Zee KJ, Endert E, et al: Interleukin-1 receptor blockade does not affect endotoxin-induced changes in plasma thyroid hormone and thyrotropin concentrations in man, J Clin Endocrinol Metab 80:1341–1346, 1995.

103.     de Metz J, Romijn JA, Endert E, et al: Administration of interferon-γ in healthy subjects does not modulate thyroid hormone metabolism, Thyroid 10:87–91, 2000.

104.     van der Poll T, Romijn JA, Wiersinga WM, et al: Tumor necrosis factor: a putative mediator of the sick euthyroid syndrome in man, J Clin Endocrinol Metab 71:1567–1572, 1990

105.     Chopra IJ, Sakane S, Chua Teco GN: A study of the serum concentration of tumor necrosis factor—in thyroidal and nonthyroidal illnesses, J Clin Endocrinol Metab 72:1113–1116, 1991.

106.     van der Poll T, Endert E, Coyle SM, et al: Neutralization of TNF does not influence endotoxin-induced changes in thyroid hormone metabolism in humans, Am J Physiol 276:R357-R362, 1999.

107.     Nagaya T, Fujieda M, Otsuka G, et al: A potential role of activated NF-kappa B in the pathogenesis of euthyroid sick syndrome, J Clin Invest 106(3):393–402, 2000.

108.     Bartalena L, Brogioni S, Grasso L, et al: Relationship of the increased serum interleukin-6 concentration to changes of thyroid function in nonthyroidal illness, J Endocrinol Invest 17:269–274, 1994.

109.     Stouthard JML, van der Poll T, Endert E, et al: Effects of acute and chronic interleukin 6 administration on thyroid hormone metabolism in humans, J Clin Endocrinol Metab 79:1342–1346, 1994.

110.    Boelen A1, Platvoet-ter Schiphorst MC, Wiersinga WM Immunoneutralization of interleukin-1, tumor necrosis factor, interleukin-6 or interferon does not prevent the LPS-induced sick euthyroid syndrome in mice. J Endocrinol. 1997 Apr;153(1):115-22.

111.     Boelen A, Platvoet-ter Schiphorst MC, Wiersinga WM: Relationship between serum 3,5,3′-triiodothyronine and serum interleukin 8, interleukin 10 or interferon gamma in patients with nonthyroidal illness, J Endocrinol Invest 19:480–483, 1996.

112.     Michalaki M, Vagenakis AG, Makri M, et al: Dissociation of the early decline in serum T3 concentration and serum IL-6 rise and TNFα in nonthyroidal illnss syndrome induced by abdominal surgery, J Clin Endocrinol Metab 86:4198–4205, 2001.

113.     Boelen A, Platvoet-ter Schiphorst MC, Wiersinga WM: Soluble cytokine receptors and the low 3,5,3′-triiodothyronine syndrome in patients with nonthyroidal disease, J Clin Endocrinol Metab 80:971–976, 1995.

114.     Schultes B, et al: Acute and prolonged effects of insulin-induced hypoglycemia on the pituitary-thyroid axis in humans, Metabolism 51:1370–1374, 2002.

115.     Pereira D, Procianoy R: Effect of perinatal asphyxia on thyroid-stimulating hormone and thyroid hormone levels, Acta Pediatr 92:339–345, 2003.

116.     Custro N, Scafidi V, Costanzo G, et al: Role of high blood glucagon in the reduction of serum levels of triiodothyronine in severe nonthyroid diseases, Minerva Endocrinol 14:221–226, 1989.

117.     Van den Berghe G, de Zegher F, Lauwers P: Dopamine and the sick euthyroid syndrome in critical illness, Clin Endocrinol 41:731–737, 1994.

118.     Legradi G, Emerson CH, Ahima RS, et al: Leptin prevents fasting-induced suppression of prothyrotropin-releasing hormone messenger ribonucleic acid in neurons of the hypothalamic paraventricular nucleus, Endocrinology 138:2569–2576, 1997.

119.     Legradi G, Emerson CH, Ahima RS, et al: Arcuate nucleus ablation prevents fasting-induced suppression of proTRH mRNA in the hypothalamic paraventricular nucleus, Neuroendocrinology 68:89–97, 1998.

120.     Flier JS, Harris M, Hollenberg AN: Leptin, nutrition, and the thyroid: the why, the wherefore, and the wiring, J Clin Invest 105:859–861, 2000.

121.     Bornstein SR, et al: Leptin levels are elevated despite low thyroid hormone levels in the “euthyroid sick” syndrome, J Clin Endocrinol Metab 82(12):4278–4279, 1997.

122.     Vesely DL, San Miguel GI, Hassan I, et al: Atrial natriuretic hormone, vessel dilator, long-acting natriuretic hormone, and kaliuretic hormone decrease the circulating concentrations of total and free T4 and free T3 with reciprocal increase in TSH, J Clin Endocrinol Metab 86:5438–5442, 2001.

123.     Kidess AI, Caplan RH, Reynertson RH, et al: Transient corticotropin deficiency in critical illness, Mayo Clin Proc 68:435–441, 1993.

124.     Merry WH, Caplan RH, Wickus GG, et al: Acute adrenal failure due to transient corticotropin deficiency in postoperative patients, Surgery 116:1095–1100, 1994.

125.     Lambert SWJ, Bruining HA, DeLong FH: Corticosteroid therapy in severe illness, N Engl J Med 337:1285–1292, 1997.

126.     Becker RA, Vaughan GM, Ziegler MG, et al: Hyper-metabolic low triiodothyronine syndrome of burn injury, Critical Care Med 10:870–875, 1982.

127.     Siegel N, et al: Beneficial effect of thyroxine on recovery from toxic acute renal failure, Kidney Int 25:906–911, 1984.

128.     Acker CG, Singh AR, Flick RP, et al: A trial of thyroxine in acute renal failure, Kidney Int 57:293–298, 2000.

129.     Acker CG, Flick R, Shapiro R, et al: Thyroid hormone in the treatment of post-transplant acute tubular necrosis (ATN), Am J Transplant 2:57–61, 2002.

130.     Little JS: Effect of thyroid hormone on survival after bacterial infection, Endocrinology 117:1431–1435, 1985.

131.     Chopra IJ, Huang TS, Boado R, et al: Evidence against benefit from replacement doses of thyroid hormones in nonthyroidal illness: studies using turpentine oil-injected rat, J Endocrinol Invest 10:559, 1987.

132.     Hsu R-B, Huang T-S, Chen Y-S, et al: Effect of triiodothyronine administration in experimental myocardial injury, J Endocrinol Invest 18:702–709, 1995.

133.     Katzeff HL, Powell SR, Ojamaa K: Alterations in cardiac contractility and gene expression during low T3 syndrome: prevention with T3, Amer J Physiol 273:E951–E956, 1997.

134.     Chapital AD, Hendrick SR, Lloyd L, et al: The effects of triiodothyronine augmentation on antithrombin III levels in sepsis, Am Surg 67:253–255, 2001.

135.     Shigematsu H, Shatney CH: The effect of triiodothyronine and reverse triiodothyronine on canine hemorrhagic shock, Nippon Geka Gakkai Zasshi 89:1587–1593, 1988.

136.     Facktor MA, Mayor GH, Nachreiner RF, et al: Thyroid hormone loss and replacement during resuscitation from cardiac arrest in dogs, Resuscitation 26:141–162, 1993.

137.     Schoenberger W, Grimm W, Emmrich P, et al: Thyroid administration lowers mortality in premature infants, Lancet 2:1181, 1979.

138      Choi YS, Kwak YL, Kim JC, Chun DH, Hong SW, Shim JK. Peri-operative oral triiodothyronine replacement therapy to prevent postoperative low triiodothyronine state following valvular heart surgery. Anaesthesia. 2009 Aug;64(8):871-7.

139.     Novitzky D, et al: Improved cardiac allograft function following triiodothyronine therapy to both donor and recipient, Transplantation 49:311–316, 1990.

140.     Kowalczuk-Wieteska A, Baranska-Kosakowska A, Zakliczynski M, Lindon S, Zembala M. Do thyroid disorders affect the postoperative course of patients in the early post-heart transplant period? Ann Transplant. 2011 Jul-Sep;16(3):77-81.

141.     Haas NA, Camphausen CK, Kececioglu D: Clinical review: thyroid hormone replacement in children after cardiac surgery: is it worth a try? Crit Care 10(3):213, 2006.

142.     Goarin JP, Cohen S, Riou B, et al: The effects of triiodothyronine on hemodynamic status and cardiac function in potential heart donors, Anesth Analg 83:41–47, 1996.

143.     Pingitore A, Galli E, Barison A, et al: Acute effects of triiodothyronine (T3) replacement therapy in patients with chronic heart failure and low-T3 syndrome: a randomized, placebo-controlled study, J Clin Endocrinol Metab 93(4):1351–1358, 2008.

144.     Spratt DI, Frohnauer M, Cyr-Alves H, et al: Physiological effects of nonthyroidal illness syndrome in patients after cardiac surgery, Am J Physiol Endocrinol Metab 293(1):E310-E315, 2007.

145.      Novitzky D1, Cooper DK2. Thyroid hormone and the stunned myocardium. J Endocrinol.        2014 Oct;223(1):R1-8. doi: 10.1530/JOE-14-0389.

146.     Hesch R, et al: Treatment of dopamine-dependent shock with triiodothyronine, Endocr Res Commun 8:299–301, 1981.

147.     Dulchavsky S, et al: T3 preserves respiratory function in sepsis, J Trauma 31:753–759, 1991.

148.     Dulchavsky S, Hendrick S, Dutta S: Pulmonary biophysical effects of triiodothyronine (T3) augmentation during sepsis-induced hypothyroidism, Trauma 35: 104–109, 1993.

149.     Bennett-Guerro E, et al: Duke T3 Study Group, Cardiovascular effects of intravenous triiodothyronine in patients undergoing coronary artery bypass graft surgery. A randomized, double-blind, placebo-controlled trial, J Am Med Assoc 275:687–692, 1996.

150      Kaptein EM, Sanchez A, Beale E, Chan LS. Clinical review: Thyroid hormone therapy for postoperative nonthyroidal illnesses: a systematic review and synthesis. J Clin Endocrinol Metab. 2010 Oct;95(10):4526-34.

151.     Hamilton MA, Stevenson LW: Thyroid hormone abnormalities in heart failure: possibilities for therapy, Thyroid 6:527–529, 1996.

152.     Kokkonen L, Majahalme S, Kööbi T, et al: Atrial fibrillation in elderly patients after cardiac surgery: postoperative hemodynamics and low postoperative serum triiodothyronine, J Cardiothorac Vasc Anesth 19(2):182–187, 2005 Apr.

153.       Weekers F, Michalaki M, Coopmans W, Van Herck E, Veldhuis JD, Darras VM, Van den Berghe G Endocrine and metabolic effects of growth hormone (GH) compared with GH-releasing peptide, thyrotropin-releasing hormone, and insulin infusion in a rabbit model of prolonged critical illness.Endocrinology. 2004 Jan;145(1):205-13.

154.       Van den Berghe G, Baxter RC, Weekers F, Wouters P, Bowers CY, Iranmanesh A,Veldhuis JD, Bouillon R: Clin Endocrinol (Oxf). 2002 May;56(5):655-69. The combined administration of GH-releasing peptide-2 (GHRP-2), TRH and GnRH tomen with prolonged critical illness evokes superior endocrine and metabolic effects compared to treatment with GHRP-2 alone

155. .      Mesotten D, Wouters PJ, Peeters RP, Hardman KV, Holly JM, Baxter RC, Van den Berghe G.: J Clin Endocrinol Metab. 2004 Jul;89(7):3105-13. Regulation of the somatotropic axis by intensive insulin therapy during protracted critical illness.

156.        Van den Berghe G, Weekers F, Baxter RC, Wouters P, Iranmanesh A, Bouillon R, Veldhuis JD. Five-day pulsatile gonadotropin-releasing hormone administration unveils combined hypothalamic-pituitary-gonadal defects underlying profound hypoandrogenism in men with prolonged critical illness. J Clin Endocrinol Metab 86:3217-3226, 2001

157.      Mebis L, Eerdekens A, Güiza F, Princen L, Derde S, Vanwijngaerden YM, Vanhorebeek I, Darras VM, Van den Berghe G, Langouche L Contribution of nutritional deficit to the pathogenesis of the nonthyroidal illness syndrome in critical illness: a rabbit model study. Endocrinology. 2012 Feb;153(2):973-84.

158.    Pérez-Guisado J, de Haro-Padilla JM, Rioja LF, Derosier LC, de la Torre JI.The potential  association of later initiation of oral/enteral nutrition on euthyroid sick syndrome in burn patients.Int J Endocrinol. 2013;2013:707360

159.      Langouche L, Vander Perre S, Marques M, Boelen A, Wouters PJ, Casaer MP, Van den Berghe G. Impact of early nutrient restriction during critical illness on the nonthyroidal illness syndrome and its relation with outcome: a randomized, controlled clinical study. J Clin Endocrinol Metab. 2013 Mar;98(3):1006-13.

160.      Herndon DN, Hart DW, Wolf SE, Chinkes DL, Wolfe RR. Reversal of catabolism by beta-blockade after severe burns. N Engl J Med 345:1223-1229, 2001.

161.      Annane D, Sebille V, Charpentier C, Bollaert P-E, Francois B, Korach J-M, Capellier G, Cohen Y, Azoulay E, Troche G, Chaumet-Riffaut P, Bellissant E. Effect of treatment with low doses of hydrocortisone and fludrocortisone on mortality in patients with septic shock. J Amer Med Assn 288:862-871, 2002.

162.      Hamrahian AH, Oseni TS, Arafah BM. Measurements of serum free cortisol in critically ill patients.N Engl J Med. 2004 Apr 15;350(16):1629-38

163.      Takala J, Ruokonen E, Webster NR, Nielsen MS, Zandstra DF, Vundelinckx G, Hinds CJ. Increased mortality associated with growth hormone treatment in critically ill adults. N Engl J Med:341:785-792, 1999. 164        Van den Berghe G. Non-thyroidal illness in the ICU: a syndrome with different faces. Thyroid. 2014 Oct;24(10):1456-65. doi: 10.1089