Archives

Cerebrotendinous Xanthomatosis

ABSTRACT

 

Cerebrotendinous xanthomatosis is a rare autosomal recessive disorder caused by homozygous or compound heterozygous mutations in the CYP27A1 gene. These patients lack mitochondrial sterol 27-hydroxylase enzyme, which is responsible for conversion of cholesterol to cholic acid and chenodeoxycholic acid (CDCA) in bile acid synthesis pathway. CYP27A1 mutation leads to decreased synthesis of bile acid, excess production of cholestanol, and consequent accumulation of cholestanol in tissues, including brain, leading to progressive neurological dysfunction marked by dementia, spinal cord paresis, and cerebellar ataxia. Deposition in other tissues causes tendon xanthomas, premature atherosclerosis, and cataracts. The clinical manifestations usually start at infancy and develop during the first and second decades of life. The diagnosis of CTX is based on clinical findings, biochemical testing, and neuroimaging. Molecular genetic analysis although not necessary for initiation of treatment, provides definitive confirmation of CTX. Early initiation of CDCA is the treatment of choice for neurological and non-neurological symptoms of CTX and treatment with cholic acid has also been shown to be effective for non-neurological symptoms.

 

INTRODUCTION

 

Cerebrotendinous xanthomatosis (CTX; OMIM#213700) is a rare autosomal recessive disorder of bile acid metabolism and lipid storage characterized by abnormal deposition of cholestanol and cholesterol in multiple tissues. It is caused by homozygous or compound heterozygous mutation in the CYP27A1 gene located on chromosome 2q33-qter. CYP27A1 gene encodes sterol 27-hydroxylase which is involved in bile acid synthesis. Over 100 different mutations (missense, deletions, insertions, splice site, and nonsense mutations) of the CYP27A1 gene have been reported worldwide in patients of different ethnic origin (1,2). About 50% of these mutations were found in the region of exons 6–8 of the CYP27A1 gene, however here is no genotype phenotype correlation (3,4). The metabolic pathway for bile acids synthesis is shown in the figure 1.

 

Figure 1. Bile acids are synthesized from cholesterol in the liver through two pathways: the classic pathway and the alternative pathway. The bile acid synthesis mainly produces two primary bile acids, cholic acid and chenodeoxycholic acid (CDCA). In the classic pathway, the mitochondrial sterol 27-hydroxylase (CYP27A1) catalyzes the steroid side-chain oxidation in both CA and CDCA synthesis. In the alternative pathway, CYP27A1 catalyzes the first step to convert cholesterol to 27-hydroxycholesterol which eventually is converted to CDCA.

 

CTX was first described by Van Bogart, Scherer and Epstein in 1937 as a condition characterized by early development of cataracts and large tendon xanthomas and later by progressive neurologic impairment (5,6). The incidence of CTX is estimated to be 3 to 5 per 100,000 people (1,7). The prevalence is estimated to be highest in Asians (1:44,407-93,084), intermediate in Europeans, Americans, and Africans/African Americans (1:70,795-233,597) and lowest in the Finnish population (1:3,388,767) (8). 

 

ETIOLOGY AND PATHOGENESIS

 

Patients with CTX lack mitochondrial sterol 27-hydroxylase, which is an important enzyme in both the alternative and classic bile acid synthesis pathways (9). The biochemical defect prevents the synthesis of chenodeoxycholic acid (CDCA) and cholic acid (10).The absence of the negative feedback mechanism of CDCA on 7a-hydroxylase, the rate limiting step, leads to the accumulation of cholestanol and its precursor 7α-hydroxy-4-cholesten-3-one (Figure 1) (9-11). Bile acid alcohols are glucoronidated and can be found in significantly increased amounts in blood, urine, and feces of untreated CTX subjects. It has been hypothesized that the increased bile alcohol levels may lead to disruption of the blood–brain barrier (BBB), and that increased permeability of BBB may be caused by circulating bile alcohol glucuronides (12). Cholestanol and cholesterol accumulate in many tissues, including brain (primarily white matter), leading to progressive neurological dysfunction marked by dementia, spinal cord paresis, and cerebellar ataxia. Deposition in other tissues causes tendon xanthomas, premature atherosclerosis, and cataracts (13).

 

CLINICAL FEATURES

 

Patients with CTX present diverse manifestations with multi-organ involvement and a broad range of neurological and non-neurological symptoms (Table 1). The clinical manifestations usually start at infancy and develop during the first and second decades of life. Neonatal cholestatic jaundice and infantile-onset diarrhea with failure to thrive may be the earliest clinical manifestation (14-16). Childhood-onset cataracts are a common early symptom, described in 92% patients with CTX. Cataracts precede neurological signs and tendon xanthoma, and if present, are considered useful for early diagnosis (14,16,17). Tendon xanthomas have been documented in 71% patients with CTX and can appear in first, second or third decade. They are common in Achilles tendon, but can also be seen in fingers, tibial tuberosity, and triceps (17,18). Pes cavus deformity has also been described (19,20). Neurological dysfunction is almost always present in patients with CTX and usually occurs in late adolescents and early adulthood. The range of symptoms are wide and include intellectual disability; dementia; psychiatric symptoms (i.e., behavioral changes, depression, agitation, hallucination, and suicide attempts); pyramidal signs (spasticity, hyperreflexia, extensor plantar responses); cerebellar signs (progressive ataxia, dysarthria); movement disorder (parkinsonism, dystonia, myoclonus, postural tremor); seizures; and peripheral neuropathy (1,4,21-23). Osteoporosis, heart involvement, and premature atherosclerosis have also been described later in life (19,24-26).

 

Table 1. Clinical Features of Cerebrotendinous Xanthomatosis

Early childhood

            Neonatal cholestatic jaundice

            Chronic diarrhea

            Cataracts

            Developmental Delay

Late childhood

            Tendon xanthomas

            Psychiatric disorders

Adulthood

            Neurological dysfunction

                        Pyramidal signs (spasticity, hyperreflexia, extensor plantar responses)

                        Cerebellar signs (ataxia, dysarthria)

                        Movement disorder (parkinsonism, dystonia, myoclonus, postural tremor)

                        Seizures

                        Dementia

            Pes Cavus

            Osteoporosis

            Premature atherosclerosis

 

DIAGNOSIS

 

The diagnosis of CTX is mainly based on clinical features, biochemical testing, neuroimaging, and molecular genetic analysis. Since clinical presentation can be variable in type, severity, and timing, diagnosis is often delayed. Patients with CTX include having high plasma cholestanol concentration (five- to ten-fold greater than normal), normal-to-low plasma cholesterol concentration, elevated urine bile alcohol concentration, elevated plasma bile alcohol concentration, decreased CDCA level, and increased levels of cholestanol and apolipoprotein B in cerebrospinal fluid (10,17). Cholesterol concentration in tissue is increased but plasma cholesterol levels are low to normal (27). Brain MRI features are strongly suggestive of diagnosis and include bilateral hyperintensity of the dentate nuclei, diffuse cerebral and cerebellar atrophy, and white matter signal abnormalities. Brain MRI spectroscopy shows decreased n-acetylaspartate and increased lactate signals, suggestive of widespread axonal damage and cerebral mitochondrial dysfunction (20). Although not necessary for initiation of treatment, molecular testing provides definitive confirmation of CTX. Various mutations in all nine exons and in introns 2,4,6,7 and 8 of the CYP27A1 gene have been described worldwide (8). Mignarri et al (22) developed a suspicion index to be used by clinicians to calculate CTX prediction score. They proposed using family history, systemic and neurologic features as diagnostic indicators, classified as very strong (score of 100; e.g., tendon xanthoma or, sibling with CTX); strong (score of 50; e.g., juvenile cataract, chronic diarrhea, prolonged neonatal cholestasis, ataxia, MRI alterations, intellectual disability and/or psychiatric disturbances); or moderate (score of 25; e.g., early osteoporosis, epilepsy, parkinsonism or polyneuropathy. A total score ≥ 100 warrants serum cholestanol assessment, and elevated cholestanol or total score ≥ 200, with one very strong or four strong indicators, warrants CYP27A1 gene analysis (22). Quantification of bile acid precursor 7α-hydroxy-4-cholesten-3-one has been proposed to be a rapid, convenient diagnostic test for CTX (28). Early detection of ketosterol bile acid precursors can play important role in early detection through newborn screening (29).

 

MANAGEMENT

 

Early initiation of oral chenodeoxy-cholic-acid (CDCA) therapy at a dose of 250 mg given 3 times daily for adults and 15 mg/kg/d for children is the treatment of choice for neurological and non-neurological symptoms such as diarrhea (30-32), however it still does not have U.S. Food and Drug Administration approval for CTX. FDA has granted it an orphan drug designation for use in CTX.  CDCA has been approved in the European Union to treat adults and children with CTX over 1 month of age. This treatment suppresses synthesis of cholesterol, cholestanol, bile alcohols, and bile acids and alleviates clinical symptoms if started at an early age (33,34); however it has not been shown to improve bone mineral density in affected patients (35). The treatment is most effective when started early (31), and patients diagnosed later in life with significant neurological disease may progress despite CDCA therapy. A retrospective cohort study in 79 Dutch patients with CTX showed that the MRI brain remained normal even after 25 year of follow-up treatment if therapy was started in young patients with a normal MRI at diagnosis. Treatment with cholic acid has also been shown to be effective, especially in patients with side effects to CDCA therapy (36,37). HMG-CoA-reductase-inhibitors along with CDCA can improve lipoprotein metabolism, inhibit cholesterol synthesis, and reduce plasma levels of cholestanol (38). Surgical excision of bilateral tendon may worsen the gait imbalance and cannot prevent the deterioration of neurologically affected patients (39). Lumbreras et al (40) showed that adeno-associated virus (AAV) vectors expressing CYP27A1 restored bile acid metabolism and normalized the concentration of most bile acids in plasma in a CTX mouse model, making gene therapy a feasible option.

 

SCREENING

 

Screening of all first-degree relatives, when feasible, should be considered as early detection and treatment are the most important aspects for preventing morbidity and mortality (41). A recent pilot study has also shown promising option of screening for CTX as part of newborn screening (42). A recent study on de-identified dried blood spots of 20,076 newborns from the 2019 cohort of the biobank of the Dutch newborn screening program (Dutch National Institute for Puublic Health and the Environment, Bilthoven, The Netherlands) showed that metabolite ratios bile alcohol cholestanetetrol glucuronide (GlcA-tetrol)/ tauro-chenodeoxycholic acid (t-CDCA) to be informative biomarker, paving way for introduction of CTX newborn screen (43).

 

CONCLUSIONS

 

Cerebrotendinous xanthomatosis is a rare autosomal recessive disorder of bile acid metabolism and lipid storage characterized by abnormal deposition of cholestanol and cholesterol in multiple tissues. Clinical manifestations include neonatal cholestatic jaundice, infantile-onset diarrhea with failure to thrive, childhood-onset cataracts, tendon xanthomas, and progressive neurological dysfunction including intellectual disability, dementia, and psychiatric symptoms. Treatment with CDCA is treatment of choice, however cholic acid, HMG-Co-A reductase inhibitors, and surgical excision also play a role in management. Timely detection and treatment are the key to prevent severe morbidity and mortality in these patients.

 

REFERENCES

 

  1. Salen G, Steiner RD. Epidemiology, diagnosis, and treatment of cerebrotendinous xanthomatosis (CTX). J Inherit Metab Dis 2017; 40:771-781
  2. Chen C, Zhang Y, Wu H, Sun YM, Cai YH, Wu JJ, Wang J, Gong LY, Ding ZT. Clinical and molecular genetic features of cerebrotendinous xanthomatosis patients in Chinese families. Metab Brain Dis 2017; 32:1609-1618
  3. Gallus GN, Dotti MT, Federico A. Clinical and molecular diagnosis of cerebrotendinous xanthomatosis with a review of the mutations in the CYP27A1 gene. Neurol Sci 2006; 27:143-149
  4. Verrips A, Hoefsloot LH, Steenbergen GC, Theelen JP, Wevers RA, Gabreels FJ, van Engelen BG, van den Heuvel LP. Clinical and molecular genetic characteristics of patients with cerebrotendinous xanthomatosis. Brain2000; 123 ( Pt 5):908-919
  5. van Bogaert L, Scherer, H.J. and Epstein, E. . Une forme cérébrale de la cholestérinose généralisée. Paris: Masson et Cie.
  6. Swanson PD. Cerebrotendinous xanthomatosis. N Engl J Med 1968; 278:857
  7. Lorincz MT, Rainier S, Thomas D, Fink JK. Cerebrotendinous xanthomatosis: possible higher prevalence than previously recognized. Arch Neurol 2005; 62:1459-1463
  8. Pramparo T, Steiner RD, Rodems S, Jenkinson C. Allelic prevalence and geographic distribution of cerebrotendinous xanthomatosis. Orphanet J Rare Dis 2023; 18:13
  9. Russell DW. The enzymes, regulation, and genetics of bile acid synthesis. Annu Rev Biochem 2003; 72:137-174
  10. Lorbek G, Lewinska M, Rozman D. Cytochrome P450s in the synthesis of cholesterol and bile acids--from mouse models to human diseases. FEBS J 2012; 279:1516-1533
  11. Bjorkhem I, Hansson M. Cerebrotendinous xanthomatosis: an inborn error in bile acid synthesis with defined mutations but still a challenge. Biochem Biophys Res Commun 2010; 396:46-49
  12. Salen G, Berginer V, Shore V, Horak I, Horak E, Tint GS, Shefer S. Increased concentrations of cholestanol and apolipoprotein B in the cerebrospinal fluid of patients with cerebrotendinous xanthomatosis. Effect of chenodeoxycholic acid. N Engl J Med 1987; 316:1233-1238
  13. Cali JJ, Hsieh CL, Francke U, Russell DW. Mutations in the bile acid biosynthetic enzyme sterol 27-hydroxylase underlie cerebrotendinous xanthomatosis. J Biol Chem 1991; 266:7779-7783
  14. Cruysberg JR. Cerebrotendinous xanthomatosis: juvenile cataract and chronic diarrhea before the onset of neurologic disease. Arch Neurol 2002; 59:1975
  15. van Heijst AF, Verrips A, Wevers RA, Cruysberg JR, Renier WO, Tolboom JJ. Treatment and follow-up of children with cerebrotendinous xanthomatosis. Eur J Pediatr 1998; 157:313-316
  16. Clayton PT, Verrips A, Sistermans E, Mann A, Mieli-Vergani G, Wevers R. Mutations in the sterol 27-hydroxylase gene (CYP27A) cause hepatitis of infancy as well as cerebrotendinous xanthomatosis. J Inherit Metab Dis 2002; 25:501-513
  17. Moghadasian MH. Cerebrotendinous xanthomatosis: clinical course, genotypes and metabolic backgrounds. Clin Invest Med 2004; 27:42-50
  18. Keren Z, Falik-Zaccai TC. Cerebrotendinous xanthomatosis (CTX): a treatable lipid storage disease. Pediatr Endocrinol Rev 2009; 7:6-11
  19. Berginer VM, Shany S, Alkalay D, Berginer J, Dekel S, Salen G, Tint GS, Gazit D. Osteoporosis and increased bone fractures in cerebrotendinous xanthomatosis. Metabolism 1993; 42:69-74
  20. Berginer VM, Abeliovich D. Genetics of cerebrotendinous xanthomatosis (CTX): an autosomal recessive trait with high gene frequency in Sephardim of Moroccan origin. Am J Med Genet 1981; 10:151-157
  21. Nie S, Chen G, Cao X, Zhang Y. Cerebrotendinous xanthomatosis: a comprehensive review of pathogenesis, clinical manifestations, diagnosis, and management. Orphanet J Rare Dis 2014; 9:179
  22. Mignarri A, Gallus GN, Dotti MT, Federico A. A suspicion index for early diagnosis and treatment of cerebrotendinous xanthomatosis. J Inherit Metab Dis 2014; 37:421-429
  23. Stelten BML, van de Warrenburg BPC, Wevers RA, Verrips A. Movement disorders in cerebrotendinous xanthomatosis. Parkinsonism Relat Disord 2019; 58:12-16
  24. Bjorkhem I, Andersson O, Diczfalusy U, Sevastik B, Xiu RJ, Duan C, Lund E. Atherosclerosis and sterol 27-hydroxylase: evidence for a role of this enzyme in elimination of cholesterol from human macrophages. Proc Natl Acad Sci U S A 1994; 91:8592-8596
  25. Valdivielso P, Calandra S, Duran JC, Garuti R, Herrera E, Gonzalez P. Coronary heart disease in a patient with cerebrotendinous xanthomatosis. J Intern Med 2004; 255:680-683
  26. Fujiyama J, Kuriyama M, Arima S, Shibata Y, Nagata K, Takenaga S, Tanaka H, Osame M. Atherogenic risk factors in cerebrotendinous xanthomatosis. Clin Chim Acta 1991; 200:1-11
  27. Salen G. Cholestanol deposition in cerebrotendinous xanthomatosis. A possible mechanism. Ann Intern Med1971; 75:843-851
  28. DeBarber AE, Connor WE, Pappu AS, Merkens LS, Steiner RD. ESI-MS/MS quantification of 7alpha-hydroxy-4-cholesten-3-one facilitates rapid, convenient diagnostic testing for cerebrotendinous xanthomatosis. Clin Chim Acta 2010; 411:43-48
  29. DeBarber AE, Luo J, Star-Weinstock M, Purkayastha S, Geraghty MT, Chiang JP, Merkens LS, Pappu AS, Steiner RD. A blood test for cerebrotendinous xanthomatosis with potential for disease detection in newborns. J Lipid Res 2014; 55:146-154
  30. Sekijima Y, Koyama S, Yoshinaga T, Koinuma M, Inaba Y. Nationwide survey on cerebrotendinous xanthomatosis in Japan. J Hum Genet 2018; 63:271-280
  31. Duell PB, Salen G, Eichler FS, DeBarber AE, Connor SL, Casaday L, Jayadev S, Kisanuki Y, Lekprasert P, Malloy MJ, Ramdhani RA, Ziajka PE, Quinn JF, Su KG, Geller AS, Diffenderfer MR, Schaefer EJ. Diagnosis, treatment, and clinical outcomes in 43 cases with cerebrotendinous xanthomatosis. Journal of clinical lipidology2018; 12:1169-1178
  32. Stelten BML, Huidekoper HH, van de Warrenburg BPC, Brilstra EH, Hollak CEM, Haak HR, Kluijtmans LAJ, Wevers RA, Verrips A. Long-term treatment effect in cerebrotendinous xanthomatosis depends on age at treatment start. Neurology 2019; 92:e83-e95
  33. Salen G, Meriwether TW, Nicolau G. Chenodeoxycholic acid inhibits increased cholesterol and cholestanol synthesis in patients with cerebrotendinous xanthomatosis. Biochem Med 1975; 14:57-74
  34. Berginer VM, Salen G, Shefer S. Long-term treatment of cerebrotendinous xanthomatosis with chenodeoxycholic acid. N Engl J Med 1984; 311:1649-1652
  35. Zubarioglu T, Bilen IP, Kiykim E, Dogan BB, Enver EO, Cansever MS, Zeybek ACA. Evaluation of the effect of chenodeoxycholic acid treatment on skeletal system findings in patients with cerebrotendinous xanthomatosis. Turk Pediatri Ars 2019; 54:113-118
  36. Koopman BJ, Wolthers BG, van der Molen JC, Waterreus RJ. Bile acid therapies applied to patients suffering from cerebrotendinous xanthomatosis. Clin Chim Acta 1985; 152:115-122
  37. Mandia D, Chaussenot A, Besson G, Lamari F, Castelnovo G, Curot J, Duval F, Giral P, Lecerf JM, Roland D, Pierdet H, Douillard C, Nadjar Y. Cholic acid as a treatment for cerebrotendinous xanthomatosis in adults. J Neurol 2019; 266:2043-2050
  38. Kuriyama M, Tokimura Y, Fujiyama J, Utatsu Y, Osame M. Treatment of cerebrotendinous xanthomatosis: effects of chenodeoxycholic acid, pravastatin, and combined use. J Neurol Sci 1994; 125:22-28
  39. Moghadasian MH, Salen G, Frohlich JJ, Scudamore CH. Cerebrotendinous xanthomatosis: a rare disease with diverse manifestations. Arch Neurol 2002; 59:527-529
  40. Lumbreras S, Ricobaraza A, Baila-Rueda L, Gonzalez-Aparicio M, Mora-Jimenez L, Uriarte I, Bunuales M, Avila MA, Monte MJ, Marin JJG, Cenarro A, Gonzalez-Aseguinolaza G, Hernandez-Alcoceba R. Gene supplementation of CYP27A1 in the liver restores bile acid metabolism in a mouse model of cerebrotendinous xanthomatosis. Mol Ther Methods Clin Dev 2021; 22:210-221
  41. Wilson DP, Patni N. Should children with chronic diarrhea be referred to a lipid clinic? Journal of clinical lipidology 2018; 12:1099-1101
  42. DeBarber AE, Kalfon L, Fedida A, Fleisher Sheffer V, Ben Haroush S, Chasnyk N, Shuster Biton E, Mandel H, Jeffries K, Shinwell ES, Falik-Zaccai TC. Newborn screening for cerebrotendinous xanthomatosis is the solution for early identification and treatment. J Lipid Res 2018; 59:2214-2222
  43. Vaz FM, Jamal Y, Barto R, Gelb MH, DeBarber AE, Wevers RA, Nelen MR, Verrips A, Bootsma AH, Bouva MJ, Kleise N, van der Zee W, He T, Salomons GS, Huidekoper HH. Newborn screening for Cerebrotendinous Xanthomatosis: A retrospective biomarker study using both flow-injection and UPLC-MS/MS analysis in 20,000 newborns. Clin Chim Acta 2023; 539:170-174

 

Cerebrotendinous Xanthomatosis

ABSTRACT

 

Cerebrotendinous xanthomatosis is a rare autosomal recessive disorder caused by homozygous or compound heterozygous mutations in the CYP27A1 gene. These patients lack mitochondrial sterol 27-hydroxylase enzyme, which is responsible for conversion of cholesterol to cholic acid and chenodeoxycholic acid (CDCA) in bile acid synthesis pathway. CYP27A1 mutation leads to decreased synthesis of bile acid, excess production of cholestanol, and consequent accumulation of cholestanol in tissues, including brain, leading to progressive neurological dysfunction marked by dementia, spinal cord paresis, and cerebellar ataxia. Deposition in other tissues causes tendon xanthomas, premature atherosclerosis, and cataracts. The clinical manifestations usually start at infancy and develop during the first and second decades of life. The diagnosis of CTX is based on clinical findings, biochemical testing, and neuroimaging. Molecular genetic analysis although not necessary for initiation of treatment, provides definitive confirmation of CTX. Early initiation of CDCA is the treatment of choice for neurological and non-neurological symptoms of CTX and treatment with cholic acid has also been shown to be effective for non-neurological symptoms.

 

INTRODUCTION

 

Cerebrotendinous xanthomatosis (CTX; OMIM#213700) is a rare autosomal recessive disorder of bile acid metabolism and lipid storage characterized by abnormal deposition of cholestanol and cholesterol in multiple tissues. It is caused by homozygous or compound heterozygous mutation in the CYP27A1 gene located on chromosome 2q33-qter. CYP27A1 gene encodes sterol 27-hydroxylase which is involved in bile acid synthesis. Over 100 different mutations (missense, deletions, insertions, splice site, and nonsense mutations) of the CYP27A1 gene have been reported worldwide in patients of different ethnic origin (1,2). About 50% of these mutations were found in the region of exons 6–8 of the CYP27A1 gene, however here is no genotype phenotype correlation (3,4). The metabolic pathway for bile acids synthesis is shown in the figure 1.

 

Figure 1. Bile acids are synthesized from cholesterol in the liver through two pathways: the classic pathway and the alternative pathway. The bile acid synthesis mainly produces two primary bile acids, cholic acid and chenodeoxycholic acid (CDCA). In the classic pathway, the mitochondrial sterol 27-hydroxylase (CYP27A1) catalyzes the steroid side-chain oxidation in both CA and CDCA synthesis. In the alternative pathway, CYP27A1 catalyzes the first step to convert cholesterol to 27-hydroxycholesterol which eventually is converted to CDCA.

 

CTX was first described by Van Bogart, Scherer and Epstein in 1937 as a condition characterized by early development of cataracts and large tendon xanthomas and later by progressive neurologic impairment (5,6). The incidence of CTX is estimated to be 3 to 5 per 100,000 people (1,7). The prevalence is estimated to be highest in Asians (1:44,407-93,084), intermediate in Europeans, Americans, and Africans/African Americans (1:70,795-233,597) and lowest in the Finnish population (1:3,388,767) (8). 

 

ETIOLOGY AND PATHOGENESIS

 

Patients with CTX lack mitochondrial sterol 27-hydroxylase, which is an important enzyme in both the alternative and classic bile acid synthesis pathways (9). The biochemical defect prevents the synthesis of chenodeoxycholic acid (CDCA) and cholic acid (10).The absence of the negative feedback mechanism of CDCA on 7a-hydroxylase, the rate limiting step, leads to the accumulation of cholestanol and its precursor 7α-hydroxy-4-cholesten-3-one (Figure 1) (9-11). Bile acid alcohols are glucoronidated and can be found in significantly increased amounts in blood, urine, and feces of untreated CTX subjects. It has been hypothesized that the increased bile alcohol levels may lead to disruption of the blood–brain barrier (BBB), and that increased permeability of BBB may be caused by circulating bile alcohol glucuronides (12). Cholestanol and cholesterol accumulate in many tissues, including brain (primarily white matter), leading to progressive neurological dysfunction marked by dementia, spinal cord paresis, and cerebellar ataxia. Deposition in other tissues causes tendon xanthomas, premature atherosclerosis, and cataracts (13).

 

CLINICAL FEATURES

 

Patients with CTX present diverse manifestations with multi-organ involvement and a broad range of neurological and non-neurological symptoms (Table 1). The clinical manifestations usually start at infancy and develop during the first and second decades of life. Neonatal cholestatic jaundice and infantile-onset diarrhea with failure to thrive may be the earliest clinical manifestation (14-16). Childhood-onset cataracts are a common early symptom, described in 92% patients with CTX. Cataracts precede neurological signs and tendon xanthoma, and if present, are considered useful for early diagnosis (14,16,17). Tendon xanthomas have been documented in 71% patients with CTX and can appear in first, second or third decade. They are common in Achilles tendon, but can also be seen in fingers, tibial tuberosity, and triceps (17,18). Pes cavus deformity has also been described (19,20). Neurological dysfunction is almost always present in patients with CTX and usually occurs in late adolescents and early adulthood. The range of symptoms are wide and include intellectual disability; dementia; psychiatric symptoms (i.e., behavioral changes, depression, agitation, hallucination, and suicide attempts); pyramidal signs (spasticity, hyperreflexia, extensor plantar responses); cerebellar signs (progressive ataxia, dysarthria); movement disorder (parkinsonism, dystonia, myoclonus, postural tremor); seizures; and peripheral neuropathy (1,4,21-23). Osteoporosis, heart involvement, and premature atherosclerosis have also been described later in life (19,24-26).

 

Table 1. Clinical Features of Cerebrotendinous Xanthomatosis

Early childhood

            Neonatal cholestatic jaundice

            Chronic diarrhea

            Cataracts

            Developmental Delay

Late childhood

            Tendon xanthomas

            Psychiatric disorders

Adulthood

            Neurological dysfunction

                        Pyramidal signs (spasticity, hyperreflexia, extensor plantar responses)

                        Cerebellar signs (ataxia, dysarthria)

                        Movement disorder (parkinsonism, dystonia, myoclonus, postural tremor)

                        Seizures

                        Dementia

            Pes Cavus

            Osteoporosis

            Premature atherosclerosis

 

DIAGNOSIS

 

The diagnosis of CTX is mainly based on clinical features, biochemical testing, neuroimaging, and molecular genetic analysis. Since clinical presentation can be variable in type, severity, and timing, diagnosis is often delayed. Patients with CTX include having high plasma cholestanol concentration (five- to ten-fold greater than normal), normal-to-low plasma cholesterol concentration, elevated urine bile alcohol concentration, elevated plasma bile alcohol concentration, decreased CDCA level, and increased levels of cholestanol and apolipoprotein B in cerebrospinal fluid (10,17). Cholesterol concentration in tissue is increased but plasma cholesterol levels are low to normal (27). Brain MRI features are strongly suggestive of diagnosis and include bilateral hyperintensity of the dentate nuclei, diffuse cerebral and cerebellar atrophy, and white matter signal abnormalities. Brain MRI spectroscopy shows decreased n-acetylaspartate and increased lactate signals, suggestive of widespread axonal damage and cerebral mitochondrial dysfunction (20). Although not necessary for initiation of treatment, molecular testing provides definitive confirmation of CTX. Various mutations in all nine exons and in introns 2,4,6,7 and 8 of the CYP27A1 gene have been described worldwide (8). Mignarri et al (22) developed a suspicion index to be used by clinicians to calculate CTX prediction score. They proposed using family history, systemic and neurologic features as diagnostic indicators, classified as very strong (score of 100; e.g., tendon xanthoma or, sibling with CTX); strong (score of 50; e.g., juvenile cataract, chronic diarrhea, prolonged neonatal cholestasis, ataxia, MRI alterations, intellectual disability and/or psychiatric disturbances); or moderate (score of 25; e.g., early osteoporosis, epilepsy, parkinsonism or polyneuropathy. A total score ≥ 100 warrants serum cholestanol assessment, and elevated cholestanol or total score ≥ 200, with one very strong or four strong indicators, warrants CYP27A1 gene analysis (22). Quantification of bile acid precursor 7α-hydroxy-4-cholesten-3-one has been proposed to be a rapid, convenient diagnostic test for CTX (28). Early detection of ketosterol bile acid precursors can play important role in early detection through newborn screening (29).

 

MANAGEMENT

 

Early initiation of oral chenodeoxy-cholic-acid (CDCA) therapy at a dose of 250 mg given 3 times daily for adults and 15 mg/kg/d for children is the treatment of choice for neurological and non-neurological symptoms such as diarrhea (30-32), however it still does not have U.S. Food and Drug Administration approval for CTX. FDA has granted it an orphan drug designation for use in CTX.  CDCA has been approved in the European Union to treat adults and children with CTX over 1 month of age. This treatment suppresses synthesis of cholesterol, cholestanol, bile alcohols, and bile acids and alleviates clinical symptoms if started at an early age (33,34); however it has not been shown to improve bone mineral density in affected patients (35). The treatment is most effective when started early (31), and patients diagnosed later in life with significant neurological disease may progress despite CDCA therapy. A retrospective cohort study in 79 Dutch patients with CTX showed that the MRI brain remained normal even after 25 year of follow-up treatment if therapy was started in young patients with a normal MRI at diagnosis. Treatment with cholic acid has also been shown to be effective, especially in patients with side effects to CDCA therapy (36,37). HMG-CoA-reductase-inhibitors along with CDCA can improve lipoprotein metabolism, inhibit cholesterol synthesis, and reduce plasma levels of cholestanol (38). Surgical excision of bilateral tendon may worsen the gait imbalance and cannot prevent the deterioration of neurologically affected patients (39). Lumbreras et al (40) showed that adeno-associated virus (AAV) vectors expressing CYP27A1 restored bile acid metabolism and normalized the concentration of most bile acids in plasma in a CTX mouse model, making gene therapy a feasible option.

 

SCREENING

 

Screening of all first-degree relatives, when feasible, should be considered as early detection and treatment are the most important aspects for preventing morbidity and mortality (41). A recent pilot study has also shown promising option of screening for CTX as part of newborn screening (42). A recent study on de-identified dried blood spots of 20,076 newborns from the 2019 cohort of the biobank of the Dutch newborn screening program (Dutch National Institute for Puublic Health and the Environment, Bilthoven, The Netherlands) showed that metabolite ratios bile alcohol cholestanetetrol glucuronide (GlcA-tetrol)/ tauro-chenodeoxycholic acid (t-CDCA) to be informative biomarker, paving way for introduction of CTX newborn screen (43).

 

CONCLUSIONS

 

Cerebrotendinous xanthomatosis is a rare autosomal recessive disorder of bile acid metabolism and lipid storage characterized by abnormal deposition of cholestanol and cholesterol in multiple tissues. Clinical manifestations include neonatal cholestatic jaundice, infantile-onset diarrhea with failure to thrive, childhood-onset cataracts, tendon xanthomas, and progressive neurological dysfunction including intellectual disability, dementia, and psychiatric symptoms. Treatment with CDCA is treatment of choice, however cholic acid, HMG-Co-A reductase inhibitors, and surgical excision also play a role in management. Timely detection and treatment are the key to prevent severe morbidity and mortality in these patients.

 

REFERENCES

 

  1. Salen G, Steiner RD. Epidemiology, diagnosis, and treatment of cerebrotendinous xanthomatosis (CTX). J Inherit Metab Dis 2017; 40:771-781
  2. Chen C, Zhang Y, Wu H, Sun YM, Cai YH, Wu JJ, Wang J, Gong LY, Ding ZT. Clinical and molecular genetic features of cerebrotendinous xanthomatosis patients in Chinese families. Metab Brain Dis 2017; 32:1609-1618
  3. Gallus GN, Dotti MT, Federico A. Clinical and molecular diagnosis of cerebrotendinous xanthomatosis with a review of the mutations in the CYP27A1 gene. Neurol Sci 2006; 27:143-149
  4. Verrips A, Hoefsloot LH, Steenbergen GC, Theelen JP, Wevers RA, Gabreels FJ, van Engelen BG, van den Heuvel LP. Clinical and molecular genetic characteristics of patients with cerebrotendinous xanthomatosis. Brain2000; 123 ( Pt 5):908-919
  5. van Bogaert L, Scherer, H.J. and Epstein, E. . Une forme cérébrale de la cholestérinose généralisée. Paris: Masson et Cie.
  6. Swanson PD. Cerebrotendinous xanthomatosis. N Engl J Med 1968; 278:857
  7. Lorincz MT, Rainier S, Thomas D, Fink JK. Cerebrotendinous xanthomatosis: possible higher prevalence than previously recognized. Arch Neurol 2005; 62:1459-1463
  8. Pramparo T, Steiner RD, Rodems S, Jenkinson C. Allelic prevalence and geographic distribution of cerebrotendinous xanthomatosis. Orphanet J Rare Dis 2023; 18:13
  9. Russell DW. The enzymes, regulation, and genetics of bile acid synthesis. Annu Rev Biochem 2003; 72:137-174
  10. Lorbek G, Lewinska M, Rozman D. Cytochrome P450s in the synthesis of cholesterol and bile acids--from mouse models to human diseases. FEBS J 2012; 279:1516-1533
  11. Bjorkhem I, Hansson M. Cerebrotendinous xanthomatosis: an inborn error in bile acid synthesis with defined mutations but still a challenge. Biochem Biophys Res Commun 2010; 396:46-49
  12. Salen G, Berginer V, Shore V, Horak I, Horak E, Tint GS, Shefer S. Increased concentrations of cholestanol and apolipoprotein B in the cerebrospinal fluid of patients with cerebrotendinous xanthomatosis. Effect of chenodeoxycholic acid. N Engl J Med 1987; 316:1233-1238
  13. Cali JJ, Hsieh CL, Francke U, Russell DW. Mutations in the bile acid biosynthetic enzyme sterol 27-hydroxylase underlie cerebrotendinous xanthomatosis. J Biol Chem 1991; 266:7779-7783
  14. Cruysberg JR. Cerebrotendinous xanthomatosis: juvenile cataract and chronic diarrhea before the onset of neurologic disease. Arch Neurol 2002; 59:1975
  15. van Heijst AF, Verrips A, Wevers RA, Cruysberg JR, Renier WO, Tolboom JJ. Treatment and follow-up of children with cerebrotendinous xanthomatosis. Eur J Pediatr 1998; 157:313-316
  16. Clayton PT, Verrips A, Sistermans E, Mann A, Mieli-Vergani G, Wevers R. Mutations in the sterol 27-hydroxylase gene (CYP27A) cause hepatitis of infancy as well as cerebrotendinous xanthomatosis. J Inherit Metab Dis 2002; 25:501-513
  17. Moghadasian MH. Cerebrotendinous xanthomatosis: clinical course, genotypes and metabolic backgrounds. Clin Invest Med 2004; 27:42-50
  18. Keren Z, Falik-Zaccai TC. Cerebrotendinous xanthomatosis (CTX): a treatable lipid storage disease. Pediatr Endocrinol Rev 2009; 7:6-11
  19. Berginer VM, Shany S, Alkalay D, Berginer J, Dekel S, Salen G, Tint GS, Gazit D. Osteoporosis and increased bone fractures in cerebrotendinous xanthomatosis. Metabolism 1993; 42:69-74
  20. Berginer VM, Abeliovich D. Genetics of cerebrotendinous xanthomatosis (CTX): an autosomal recessive trait with high gene frequency in Sephardim of Moroccan origin. Am J Med Genet 1981; 10:151-157
  21. Nie S, Chen G, Cao X, Zhang Y. Cerebrotendinous xanthomatosis: a comprehensive review of pathogenesis, clinical manifestations, diagnosis, and management. Orphanet J Rare Dis 2014; 9:179
  22. Mignarri A, Gallus GN, Dotti MT, Federico A. A suspicion index for early diagnosis and treatment of cerebrotendinous xanthomatosis. J Inherit Metab Dis 2014; 37:421-429
  23. Stelten BML, van de Warrenburg BPC, Wevers RA, Verrips A. Movement disorders in cerebrotendinous xanthomatosis. Parkinsonism Relat Disord 2019; 58:12-16
  24. Bjorkhem I, Andersson O, Diczfalusy U, Sevastik B, Xiu RJ, Duan C, Lund E. Atherosclerosis and sterol 27-hydroxylase: evidence for a role of this enzyme in elimination of cholesterol from human macrophages. Proc Natl Acad Sci U S A 1994; 91:8592-8596
  25. Valdivielso P, Calandra S, Duran JC, Garuti R, Herrera E, Gonzalez P. Coronary heart disease in a patient with cerebrotendinous xanthomatosis. J Intern Med 2004; 255:680-683
  26. Fujiyama J, Kuriyama M, Arima S, Shibata Y, Nagata K, Takenaga S, Tanaka H, Osame M. Atherogenic risk factors in cerebrotendinous xanthomatosis. Clin Chim Acta 1991; 200:1-11
  27. Salen G. Cholestanol deposition in cerebrotendinous xanthomatosis. A possible mechanism. Ann Intern Med1971; 75:843-851
  28. DeBarber AE, Connor WE, Pappu AS, Merkens LS, Steiner RD. ESI-MS/MS quantification of 7alpha-hydroxy-4-cholesten-3-one facilitates rapid, convenient diagnostic testing for cerebrotendinous xanthomatosis. Clin Chim Acta 2010; 411:43-48
  29. DeBarber AE, Luo J, Star-Weinstock M, Purkayastha S, Geraghty MT, Chiang JP, Merkens LS, Pappu AS, Steiner RD. A blood test for cerebrotendinous xanthomatosis with potential for disease detection in newborns. J Lipid Res 2014; 55:146-154
  30. Sekijima Y, Koyama S, Yoshinaga T, Koinuma M, Inaba Y. Nationwide survey on cerebrotendinous xanthomatosis in Japan. J Hum Genet 2018; 63:271-280
  31. Duell PB, Salen G, Eichler FS, DeBarber AE, Connor SL, Casaday L, Jayadev S, Kisanuki Y, Lekprasert P, Malloy MJ, Ramdhani RA, Ziajka PE, Quinn JF, Su KG, Geller AS, Diffenderfer MR, Schaefer EJ. Diagnosis, treatment, and clinical outcomes in 43 cases with cerebrotendinous xanthomatosis. Journal of clinical lipidology2018; 12:1169-1178
  32. Stelten BML, Huidekoper HH, van de Warrenburg BPC, Brilstra EH, Hollak CEM, Haak HR, Kluijtmans LAJ, Wevers RA, Verrips A. Long-term treatment effect in cerebrotendinous xanthomatosis depends on age at treatment start. Neurology 2019; 92:e83-e95
  33. Salen G, Meriwether TW, Nicolau G. Chenodeoxycholic acid inhibits increased cholesterol and cholestanol synthesis in patients with cerebrotendinous xanthomatosis. Biochem Med 1975; 14:57-74
  34. Berginer VM, Salen G, Shefer S. Long-term treatment of cerebrotendinous xanthomatosis with chenodeoxycholic acid. N Engl J Med 1984; 311:1649-1652
  35. Zubarioglu T, Bilen IP, Kiykim E, Dogan BB, Enver EO, Cansever MS, Zeybek ACA. Evaluation of the effect of chenodeoxycholic acid treatment on skeletal system findings in patients with cerebrotendinous xanthomatosis. Turk Pediatri Ars 2019; 54:113-118
  36. Koopman BJ, Wolthers BG, van der Molen JC, Waterreus RJ. Bile acid therapies applied to patients suffering from cerebrotendinous xanthomatosis. Clin Chim Acta 1985; 152:115-122
  37. Mandia D, Chaussenot A, Besson G, Lamari F, Castelnovo G, Curot J, Duval F, Giral P, Lecerf JM, Roland D, Pierdet H, Douillard C, Nadjar Y. Cholic acid as a treatment for cerebrotendinous xanthomatosis in adults. J Neurol 2019; 266:2043-2050
  38. Kuriyama M, Tokimura Y, Fujiyama J, Utatsu Y, Osame M. Treatment of cerebrotendinous xanthomatosis: effects of chenodeoxycholic acid, pravastatin, and combined use. J Neurol Sci 1994; 125:22-28
  39. Moghadasian MH, Salen G, Frohlich JJ, Scudamore CH. Cerebrotendinous xanthomatosis: a rare disease with diverse manifestations. Arch Neurol 2002; 59:527-529
  40. Lumbreras S, Ricobaraza A, Baila-Rueda L, Gonzalez-Aparicio M, Mora-Jimenez L, Uriarte I, Bunuales M, Avila MA, Monte MJ, Marin JJG, Cenarro A, Gonzalez-Aseguinolaza G, Hernandez-Alcoceba R. Gene supplementation of CYP27A1 in the liver restores bile acid metabolism in a mouse model of cerebrotendinous xanthomatosis. Mol Ther Methods Clin Dev 2021; 22:210-221
  41. Wilson DP, Patni N. Should children with chronic diarrhea be referred to a lipid clinic? Journal of clinical lipidology 2018; 12:1099-1101
  42. DeBarber AE, Kalfon L, Fedida A, Fleisher Sheffer V, Ben Haroush S, Chasnyk N, Shuster Biton E, Mandel H, Jeffries K, Shinwell ES, Falik-Zaccai TC. Newborn screening for cerebrotendinous xanthomatosis is the solution for early identification and treatment. J Lipid Res 2018; 59:2214-2222
  43. Vaz FM, Jamal Y, Barto R, Gelb MH, DeBarber AE, Wevers RA, Nelen MR, Verrips A, Bootsma AH, Bouva MJ, Kleise N, van der Zee W, He T, Salomons GS, Huidekoper HH. Newborn screening for Cerebrotendinous Xanthomatosis: A retrospective biomarker study using both flow-injection and UPLC-MS/MS analysis in 20,000 newborns. Clin Chim Acta 2023; 539:170-174

 

Defects of Thyroid Hormone Transport in Serum

ABSTRACT

 

Inherited abnormalities of thyroid hormone-binding proteins are not uncommon and can predominate in some ethnic groups. They alter the number of iodothyronines present in serum and, although the concentration of free hormones remains unaltered, routine measurement can give erroneous results. With a single exception, inherited defects in thyroxine-binding globulin (TBG), are X-chromosome linked and thus, the full phenotype is expressed mostly in males. Partial TBG deficiency is more common than complete deficiency. High frequency of variants TBGs have been identified in African Blacks, Australian Aborigine, and Eskimos. Most defects producing TBG deficiency are caused by mutations in the structural gene. However, inherited X-linked partial deficiency can occur as the consequence of mutations of a gene enhancer. Inherited forms of TBG excess are all caused by gene duplication or triplication. Mutations in the transthyretin (TTR) gene producing a molecule with increased affinity for T4 are relatively rare. A variant TTR produces transient hyperthyroxinemia during non-thyroidal illness.     Mutations of the human serum albumin (HSA) gene produce increased concentration of serum T4, a condition known as familial dysalbuminemic hyperthyroxinemia (FDH). They are relatively more common in individuals of Hispanic origin. They cause an increase in serum T4 owing to increased affinity for this iodothyronine but high concentrations in free T4 observed in direct measurement by some commercial methods are erroneous. A variant with increased affinity for T3 has been also identified.

 

INTRODUCTION

 

Abnormalities in the serum proteins that transport thyroid hormone do not alter the metabolic state and do not cause thyroid disease.  However, they do produce alterations in thyroid hormone concentration in serum and when unrecognized have led to inappropriate treatment.  When the abnormality is the consequence of altered synthesis, secretion or stability of the variant serum protein, the free thyroid hormone level estimated by most of the clinically available techniques remains within the range of normal. In contrast, when the defect results in a significant alteration of the affinity of the variant protein for the hormone, estimates of the free thyroid hormone level often give erroneous results and thus, it is prudent to measure the free hormone concentration by more direct methods such as equilibrium dialysis or ultrafiltration.  This is also true in cases of complete TBG deficiency, in whom the estimation of free thyroid hormone level in serum by indirect methods, or using iodothyronine analogs as tracers, can also give erroneous results.

 

The existence of inherited defects of serum transport of thyroid hormone was first recognized in 1959 with the report of TBG-excess by Beierwaltes and Robbins (1).  Genetic variants for each of the three major thyroid hormone transport proteins have since been described and in subsequent years, the molecular basis of a number of these defects has been identified (2).  Clinically, these defects usually manifest as either euthyroid hyperthyroxinemia or hypothyroxinemia and more rarely, isolated hypertriiodothyroninemia (3).  Associated abnormalities such as thyrotoxicosis, hypothyroidism, goiter, and familial hyperlipidemia are usually coincidental (4).  However, individuals with thyroid disorders are more likely to undergo thyroid testing leading to the fortuitous detection of a thyroid hormone transport defect.

 

THYROXINE-BINDING GLOBULIN (TBG) DEFECTS

 

Familial TBG abnormalities are inherited as X-chromosome linked traits (5, 6), compatible with the location of the TBG (SERPINA7) gene on the long arm of the X-chromosome (Xq22.2) (7, 8).  This mode of inheritance also suggests that the defects involve the TBG gene proper, rather than the rate of TBG disposal, as long ago postulated (5). The normal, common type TBG (TBG-N or TBG-C), has a high affinity for iodothyronines [affinity constants (Ka): 10-10 M-1 for T4 and 10-9 M-1 for T3] and binds 75% of the total T4 and T3 circulating in blood. Thus, with a single exception [HSA R218P and R218S (9-11), see below], among the inherited abnormalities of thyroid hormone transport proteins, those involving the TBG molecule produce usually more profound alterations of thyroid hormone concentration in serum.

 

Clinically TBG defects are classified according to the level of TBG in serum of affected hemizygotes (XY males or XO females, that express only the mutant allele): complete TBG deficiency (TBG-CD), partial TBG deficiency (TBG-PD) and TBG excess (TBG-E).  In families with TBG-CD, affected males have no detectable TBG and carrier females (mothers or daughters) have on the average half the normal TBG concentration (4).  In families with partially TBG deficient males, the mean TBG concentration in heterozygous females is usually above half the normal.  Serum TBG concentration in males with TBG-E is 2 to 4-fold the normal mean and that in the corresponding carrier females, is slightly higher than half that of the affected males.  These observations indicate an equal contribution of cells expressing the normal and mutant TBG genes. On rare occasions, selective inactivation of the X-chromosome has been the cause of manifestations of the complete defect (hemizygous phenotype) in heterozygous females (12, 13).

 

Inherited TBG defects can be further characterized by the level of denatured TBG (dnTBG) in serum and the physicochemical properties of the molecule.  The latter can be easily determined without the need of purification. These properties are: (a) immunologic identity; (b) isoelectric focusing (IEF) pattern; (c) rate of inactivation when exposed to various temperatures and pH; and (d) affinity for the ligands, T4 and T3.  More precise identification of TBG defects requires sequencing of the TBG gene.

 

MiP a Subject With TBG-CD

 

The proposita, a phenotypic female, was 13 years old when first seen because of retarded growth, amenorrhea and absence of secondary sexual traits.  She was the first sibling of a second marriage for both parents. The family included a younger brother and four older half-siblings, two maternal and two paternal. The proposita was born to her 30-year-old mother after full-term, uncomplicated pregnancy. Infancy and early childhood development were normal until 4 years of age when it became apparent that she was shorter than her peers. She was 12 years of age when a low protein bound iodine (PBI, then a measure of T4) of 2.2 µg/dl (normal range 4.0-8.0) was noted and treatment with 120 mg of desiccated thyroid (equivalent to 200µg L-T4) daily was initiated. Since, during the ensuing 6 months, no change in her growth rate occurred and because PBI remained unchanged (2.0 µg/dl), the dose of desiccated thyroid was increased to 180 mg/day. This produced restlessness, perturbed sleep and deterioration of school performance necessitating discontinuation of thyroid hormone treatment. No family history of thyroid disease or short stature was elicited and the parents’ denied consanguinity.

 

On physical examination, the patient appeared younger than her chronological age, was short (137 cm) and showed no signs of sexual development.  She had a webbed neck, low nuchal hairline, bilateral eyelid ptosis, shield-shaped chest, increased carrying angle and short 4th metacarpals and metatarsals. The thyroid gland was normal in size and consistency.

 

Buccal smear was negative for Barr bodies and karyotyping revealed 45 chromosomes consistent with XO Turner's syndrome. No chromosomal abnormalities were found in lymphocytes from the mother and father. Bone age was 12 years and X-ray of the scull showed a mild degree of hypertelorism. PBI and butanol extractable iodine were low at 2.0 and 1.8 µg/dl, respectively. Resin-T3 uptake was high at 59.9% (normal range 25-35%) indicating reduced TBG-binding capacity. A 24-hour thyroidal radioiodide uptake was normal at 29%, basal metabolic rate was +20% (normal range -10 to +20) and TG autoantibodies were not present.  Serum cortisol was normal as were the responses to ACTH and metyrapone.  Basal growth hormone concentration was normal at 8.0 ng/ml which rose to 32 ng/ml following insulin hypoglycemia.

 

Studies were carried out in all first-degree relatives and the proposita was treated cyclically with diethylstilbestrol which produced withdrawal uterine bleeding and gradual breast development.

 

Five family members, in addition to the proposita had thyroid function tests abnormalities. Two were males and three females.  The two males (maternal grandfather and maternal half-brother) and the proposita had the lowest PBI levels and undetectable T4-binding to serum TBG.  In contrast, the three females (mother, maternal aunt and maternal half-sister) had a lesser reduction of their PBI and T4-binding capacity to TBG approximately one-half the normal mean value. The two sons of the affected grandfather (maternal uncles to the proposita) had normal PBI and T4-binding to TBG. No interference with T4-binding to TBG or other serum protein abnormalities were found in affected members of the family. In vivo T4 kinetic studies revealed a rapid extrathyroidal turnover rate but normal daily secretion and degradation, compatible with their eumetabolic state.

 

INTERPRETATION

 

The incidental identification of thyroid tests abnormalities in the proposita is typical for most subjects with TBG deficiency as well as TBG excess. So is the initial unnecessary treatment; though less frequent with the routine measurement or estimation of free T4. The inherited nature of the defect is suspected by exclusion of factors known to cause acquired TBG abnormalities and should be confirmed by the presence of similar abnormalities in members of the family. The absence of male-to-male transmission and the carrier state of all female offspring of the affected males is a typical pattern of X-chromosome linked inheritance. This is further supported by the complete TBG deficiency in individuals having a single X chromosome (males and the XO female) and only partial TBG deficiency in carrier XX females.

 

Since the publication of this family in 1968 (14), the cause of the TBG defect was identified. From the mutation identified in the TBG gene of this family [TBG Harwichport (TBG-CD H)], it can be deduced that the molecule is truncated, missing 12 amino acids at the carboxyl terminus (15).

 

Fifty six TBG variants have been so far identified and in 50 the precise defect has been determined by gene analysis. Their primary structure defect, some of their physical and chemical properties and the resulting serum T4 concentrations are summarized in Table 1 and figure 1.

 

Figure 1. Properties of some TBG variants causing partial TBG deficiency (TBG-PD). The TBG variants are: -SD, San Diego; -G, Gary; -M, Montreal, -S, slow; -A, Aborigine; -Poly, polymorphic; -Cgo, Chicago; and -Q, Quebec. For detailed description, see (1) Sarne et al (39) and Bertenshaw et al (37); (2) Murata et al (34), Mori et al (43) and Kambe et al (60); (3) Takamatsu et al (44) and Janssen et al (45); (4) Takamatsu et al (55) and Waltz et al (56); (5) Murata et al (61) and Takeda et al (47); (6) Mori et al (26) and Takeda et al (57); (7) Takamatsu et al (59) and (58); (8) Takamatsu et al (44) and Bertenshaw et al (49). [Modified from Refetoff et al (62)].

 

 

Table 1.  TBG Variants and Gene Mutations

TBG NAME

Abbreviated

name

Intron

Exon

CODON*

AMINO ACID

NUCLEOTIDE

References

WT

Variant

WT

Variant

Complete Deficiency (CD)

Milano (fam A)

CDMi†

IVS 1

fs

5' DSS

unknown

gtaagt

gttaagt

(16)

Andrews

CDAN

IVS 1

fs

5' DSS

unknown

gtaagt

gcaagt

(17)

Portuguese 1 (pt A)

CDP1

1

23

S (Ser)

X (OCH)

TCA

TAA

(18)

Yonago

CDY

1

28-29fs-51

D F

X (OPA)

GA(CT)TT

GAATT

(19)

Negev (Bedouin

CDN

1

38fs-51

T (Thr)

X (OPA)

ACT

T del

(20, 21)

Nikita (fam B)

CDNi

1

50fs-51

P (Pro)

X (OPA)

CCT

T del

(16)

Taiwanese 1

CDT1†

1

52

S (Ser)

N (Asn)

AGC

AAC

(22)

Parana

CDPa†

1

61

S (Ser)

C (Cys)

TCC

TGC

a

No name

CD6

1

165fs-168

V (Val)

X (OCH)

GTT

T del

(23)

Kankakee

CDK

IVS 2

188fs-195

3' ASS

X (OPA)

agCC

ggCC

(24)

Poland

CDPL

2

201fs-206

D (Asp)

X (OCH)

GAC

G del

(25)

Portuguese 2(pt B)

CDP2

2

223

Q (Gln)

X (OCH)

CAA

TAA

(18)

No name

CD5†

2

227

L (Leu)

P (Pro)

CTA

CCA

(26)

Portuguese 3§

CDP3

2

233

N (Asn)

I (Ile)

ACC

ATC

(27)

Berlin

CDBn

IVS 3+3

fs

intronic

unknown

intronic

79nt del

(17)

Houston

CDH

IVS 3

279fs-374

3’ ASS

X (OPA)

agAT

aaAT

(28)

Buffalo

CDB

3

280

W (Trp)

X (AMB)

TGG

TAG

(29)

Taiwanese 2

CDT2

3

280

W (Trp)

X (OPA)

TGG

TGA

(22)

Lisle

CDL

IVS 4

280fs-325

5' DSS

X (OPA)

gtaaa

ggaaa

b

Jackson (fam K)

CDJa

IVS 4

280fs-325

5' DSS

X (OPA)

gtaaa

gtaag

(30)

No name

CD7

3

283fs-301

L (Leu)

X (OPA)

TGT

G del

(15)

No name

CD8†

4

329fs-374

A (Ala)

X (OPA)

GCT

G del

(15)

Japan

CDJ

4

352fs-374

L (Leu)

X (OPA)

CTT

C del

(31, 32)

Penapolis

CDPe

4

332fs-374

K (Lys)

X (OPA)

AAG

A del

a

Kyoto§

CDKo

4

370

S (Ser)

F (Phe)

TCT

TTT

(33)

Harwichport

CDH

4

381fs-396

Y (Tyr)

X (OPA)

AGG

19nt del

(14, 15, 34)

NeuIsenburg

CDNl

4

384fs-402

L (Leu)

7 aa add

CTC

TC del

(35)

Partial Deficiency (PD)

Allentown

PDAT

1

-2

H (His)

Y (Tyr)

CAC

TAC

(36)

San Diego

PDSD†

1

23

S (Ser)

T (Thr)

TCA

ACA

(37-39)

Brasilia

PDB

1

35

R (Arg)

W (Trp)

CGG

TGG

(40)

Wanne-Eickel

PDWE

1

35

R (Arg)

E (Glu)

CGG

CAG

(41)

No name

 

1

35

R (Arg)

Q (Gln)

CGG

TGG

(41)

No name

?

1

52

S (Ser)

R (Arg)

AGT

AGA

c

No name

?

1

64

A (Ala)

D (Asp)

GCC

GAC

(42)

Korea

PDKa

1

74

E (Glu)

K (Lys)

GAG

AAG

a

Gary

PDG

1

96

I (Ile)

N (Asn)

ATC

AAC

(43)

No name

 

1

112

N (Asn)

L (Lys)

AAT

AAG

c

Montréal

PDM

1

113

A (Ala)

P (Pro)

GCC

CCC

(44, 45)

Berlin2

Bn2

1

161

T (Thr)

N (Asn)

ACC

AAC

a.

Aborigine

PDA†

2

191

A (Ala)

T (Thr)

GCA

ACA

(46, 47)

Glencoe

PDGe

2

215

V (Val)

G (Gly)

GTG

GGG

(48)

Quebec

PDQ†

4

331

H (His)

Y (Tyr)

CAT

TAT

(44, 49)

Japan (Kumamoto)

PDJ

4

363

P (Pro)

L (Leu)

CCT

CTT

(50, 51)

Heidelberg

PDHg

4

368

D (Asp)

G (Gly)

GAT

GGT

(52)

No name

?

4

381

R (Arg)

G (Gly)

AGG

GGG

c

No name

?

4

382

S (Ser)

R (Arg)

AGT

CGT

c

No name

 

enhancer

 

-

-

G

A

(53)

Other Variants

Slow

S

1

171

D (Asp)

N (Asn)

GAC

AAC

(54-56)

Polymorphism

Poly

3

283

L (Leu)

F (Phe)

TTG

TTT

(26, 57)

Chicago

CH or Cgo

3

309

Y (Tyr)

F (Phe)

TAT

TTT

(58, 59)

* Codon numbering from fist amino acid of the mature protein. The 20 amino acids of the signal peptide are numbered -1 to -20, from N- to C-terminus.  The codon at the site of mutation is followed by the codon at the site of termination of translation.

† Coexistence of TBG Poly

  • complete deficiency is uncertain as the TBG assay used was unable to detect values <10% the mean normal

¶ Also a silent mutation at codon 55:  GCA -> GCG

a Personal communication

b Personal observation

c Communicated by Pia Hermanns and Joachim Pohlenz, University of Mainz, Germany

del, delete; add, addition; aa, amino acid; fs, frame shift

Pt, patent; fam, family

IVS, intervening sequence or intron; ASS, acceptor splice site; DSS, donor splice site

 

Complete Deficiency of TBG (TBG-CD)

 

TBG-CD is defined as undetectable TBG in serum of affected hemizygous subjects or a value lesser than 0.03% the normal mean; the current limits of detection using the most sensitive radioimmunoassay (RIA) being 5ng/dl (26).  The prevalence is approximately 1:15,000 newborn males.  Twenty-seven TBG variants having this phenotype have been characterized at the gene level.  These are shown in table 1 that also contains references to the original publications. Twenty two of the 27 TBG-CDs have truncated molecules. Early termination of translation of these variants is caused in 4 by a single nucleotide substitution (TBG-CDP1, TBG-CDP2, CD5, TBG-CDB and TBG-CDT2) or by a frame shift due to one nucleotide deletion (TBG-CDY, TBG-CDN, TBG-CDNi, TBG-CD6, CD-PL, TBG-CD7, TBG-CD8, and TBG-CDJ, TBG-CDPe) or deletion of 19 nucleotides (TBG-CDH).  In 7 variants mutations occurred in introns, 6 of which are close to splice sites (TBG-CDMi, TBG-CDAN, TBG-CDK, TBGBn, TBG-CDH, TBG-CDL and TBG-CDJa).  A mutation at the acceptor splice junction caused also a frame shift producing early termination of translation in TBG-CDK (24).  In contrast a nucleotide substitutions in the 5' donor splice site of intron IV (TBG-CDL and TBG-CDJa), resulted in a complete splicing of exon 3, also producing a truncated molecule (30) and personal observation.  A similar mechanism is likely responsible for CD in TBG-CDMi, though direct experimental prove was not provided (16).  Single amino acid substitution was the cause of CD in five families (TBG-CDT1, TBG-CDPa, TBG-CD5, TBG-CDP3 and TBG-CDKo).  In TBG-CD5 Leucine-227 with a proline was shown to cause aberrant post-translational processing (45).  One TBG variant (TBG-CDNI), with two nucleotides deleted close to the carboxyl terminus, the resulting frame shift predicts an extension of the molecule by the addition of 7 nonsense residues (35).  TBG-CDJ has been so far identified only in Japanese but its allele frequency in the population remains unknown (32, 57) (Table. 1).

 

Partial Deficiency of TBG (TBG-PD)

 

This is the most common form of inherited TBG deficiency having a prevalence of 1:4,000 newborn.  Identification of heterozygous females by serum TBG measurement may be difficult because levels often overlap the normal range.  In contrast to variants with complete TBG deficiency, all TBG-PDs have missense mutations. It is possible that three of the five variants with single amino acid substitutions included in the category of TBG-CD have also partial deficiency which was not identified owing to the low sensitivity of routine assays for the measurement of TBG.  Twenty-one different mutations, producing a variable degree of reduction of TBG concentration in serum, have been identified, 20 of which involve mutations in the TBG gene proper.  They are listed in table 1.  In addition, some of these variants are unstable (TBG-PDG, TBG-PDA, TBG-PDSD, TBG-PDM TBG-PDQ and TBG-PDJ) or have lower binding affinity for T4 and T3 (TBG-PDG, TBG-PDA, TBG-PDS TBG-PDSD, TBG-PDM and TBG-PDQ), impaired intracellular transport and secretion (TBG-PDJ and TBG-CDJ) and some exhibit an abnormal migration pattern on IEF electrophoresis (TBG-PDG, TBG-PDM, and TBG-PDQ) (Fig. 1).  Variants with decreased affinity for T4 and T3 have a disproportionate reduction in hormone concentration relative to the corresponding serum TBG level (Fig. 2) and estimations of the free hormone levels by any of the index methods gives erroneous results (39, 64).  One of these variants, TBG-PDA, is found with high frequency (allele frequency of 51%) in Australian Aborigines (47).

 

Figure 2. Serum T4-bound to TBG and the concentration of TBG and denatured TBG (dnTBG) in hemizygous subjects expressing the different TBG variants. Results, graphed as mean ± SD, were normalized by expressing them as % of those for the common type TBG (TBG-C). For abbreviations used in the nomenclature of the TBG variants, see legend to figure 1. [Adapted from Janssen et al (63)].

 

A unique family with TBG-PD has been described in which inheritance of the partial deficiency was autosomal dominant with transmission of the phenotype from father to son (65).  The concentration of TBG in affected males and females was about one half the normal mean value.  The TBG had normal affinity for T4. normal IEF and heat lability.  No sequence changes were found in the entire coding arias of the gene or in the promoter region.  Although the mechanism of TBG-CD in this family is unknown an abnormality in one of the factors regulating TBG gene transcription is a distinct possibility. Further studies to determine the genetic defect have been hampered by lack of subjects’ cooperation.

 

In 5% (4 or 74) families with X-chromosome lined TBG deficiency, studied in the author’s laboratory, no mutations were identified in the entire TBG gene, including all exons, introns, untranslated regions and the promoter region of the gene, covering a total of 9.2 kb. Next-generation sequencing identified a novel single nucleotide substitution 20 kb downstream of the TBG gene in all four families. In silico analysis predicted that the variant resides within a liver-specific enhancer. In vitro studies confirmed the enhancer activity of a 2.2-kb fragment of genomic DNA containing the novel variant and showed that the mutation reduces the activity of this enhancer. The affected subjects share a haplotype of 8 Mb surrounding the mutation. Three were of known Arab ethnicity and in all four families the most recent common ancestor was estimated to be 19.5 generations ago (95% confidence intervals). This is first report of an inherited endocrine disorder caused by a mutation in an enhancer region (53).

 

TBG Excess (TBG-E)

 

TBG-E has a lower prevalence than TBG deficiency, with values obtained from neonatal screening programs from 1:6,000 to 1:40,000 (66, 67).  Considering that some newborn may have non-inherited, transient TBG excess, a conservative overall estimate of inherited TBG-E would be 1:25,000 (68).  Early sequencing of the coding and promoter regions of subjects with TBG-E failed to show any defects (69).  However, in 1995, Mori et al (70) found that gene amplification was the cause of TBG-E in two families.  Gene triplication and duplication were demonstrated by gene dosage studies using HPLC measurements of the PCR -amplified product.  As expected, hemizygous affected males had approximately 3- and 2-fold the average normal serum TBG concentration, respectively.  The presence of multiple TBG gene copies in tandem was confirmed by in situ hybridization of prometaphase and interphase chromosomes from an affected male (Fig. 3).

Figure 3. Hybridization in prometaphase chromosomes of cultured skin fibroblasts obtained from an affected male with TBG-E. A complete TBG cDNA was used as a probe. Three TBG gene copies are seen in tandem with each exon clearly identified in the starched chromosome shown in panel B.

 

 

TBG Variants with Unaltered TBG Concentrations in Serum

 

Five TBG variants have been identified that present with normal or slight and clinically insignificant alterations in their concentration in serum.  Four occur with high frequency in some population groups and thus, can be considered as polymorphic.  TBG-Poly (Fig. 1), with no alterations of its physical or biological properties, has been detected in 16% and 20% of the French Canadian and Japanese populations, respectively (26, 57).  TBG-S exhibits a slower mobility on polyacrylamide gel electrophoresis and cathodal shift on IEF (54, 55), owing to the loss of a negative change due to the replacement of the normal Asp171 by Asn (56) (Figs. 1 and 4). It has an allele frequency of 5 to 16% in Black populations of African origin and 2 to 10% in Pacific Islanders.  The molecular structure of two other polymorphic TBG variants has not been identified.  TBG-F has an allele frequency of 3.2% in Eskimos residing on the Kodiac and St. Lawrence islands.  It has a slight anodal (fast) mobility on IEF (71).  TBG-C1 has been identified in subjects inhabiting two Mali village (72).  It has a small cathodal shift on IEF and an allele frequency of 5.1%.  TBG-Cgo, resistant to high temperatures (59), has normal affinity for T4 and T3.  All SERPINs except human TBG have a Phe at a position corresponding to Tyr309.  Structure modeling suggests that the replacement of the normal Tyr309 by Phe in TBG-Cgo, ties the internal α-helix hI1 to the molecule, thus stabilizing its tertiary structure (58).  Studies using recombinant TBG–Cgo showed that the molecule exists in loop expelled conformation.  However, when exposed at 37°C, the protein readily converts to a more stable loop inserted conformation explaining its subsequent  enhanced heat stability, as observed in vivo (73).

 

Figure 4. Microheterogeneity of TBG. Tracer amounts of l25I T4 were added to serum prior to submission to isoelectric focusing and radioautography. TBG C (common type) exhibits 6 bands spanning from pH 4.18 to 4.58. Three of the six are major and shown here between pH 4.35 and 4.50. TBG-Slow (TBG S) from a hemizygous male shows a cathodally shifted pattern. A mixed pattern occurs in heterozygous females expressing both TBG-C and TBG-S. [Reproduced from Waltz et al (56)].

 

Biological Consequences of Structural Changes Caused by Mutations in the TBG Gene

 

The mechanisms whereby structural abnormalities of the TBG molecule produce the variant phenotypes have been investigated by expression of some of these molecules in living cells.  Contrary to earlier speculation, increased extracellular degradation due to instability is a rare cause reduced concentration of the variant TBG in serum (38).  More commonly, intracellular retention and degradation of the defective TBG molecules is responsible for their presence in low concentrations in serum (45, 51, 60, 74).  Of note is the full intracellular retention of TBG-CD5 despite synthesis in normal quantities.  A single amino acid substitution in TBG-CD5 is sufficient to alter its tertiary structure and thus prevent export.  The same finding in the case of TBG-CDJ has been traced to its retention within the endoplasmic reticulum.  Furthermore, the increased amount of GRP78 mRNA in cells transfected with TBG-PDJ suggests that association of this TBG variant with the GRP78 molecular chaperon is responsible for its impaired secretion (51).  The variant TBG-AL is unique and important as it provides information about the function of the signal peptide. The resulting variable decrease in the serum TBG concentration associated with diminished in vitro secretion is compatible with impaired cotranslational processing (36). 

 

Several speculations regarding the properties of variant TBGs have been confirmed based on the elucidation of the TBG structure by X-ray crystallography (75). The reduced ligand-binding of TBG-SD (38, 39) can be explained by the direct proximity of the amino acid substitution to the binding pocket.  Indeed, the methyl group of the side chain of Thr23, replacing the normal Ser, will sterically hinder the binding of T4.  Similarly, in TBG-A, the replacement of Ala191 by Thr (47) perturbs the H-bounds that stabilizes the binding pocket, leading to the reduced T4 binding.  In contrast, the loss of His331 in TBG-Q (H331Y) (44, 49) allows unrestricted loop insertion in the upper half of the A-sheet, accounting for the increased in serum denatured (dn)TBG and reduced T4 binding.

 

TBG deficiency was found to coexist in the same family with resistance to thyroid hormone beta (RTHß) (76). Both TBG (P50fs51X) and THRB (P453A) gene mutations have been previously described in unrelated families (16, 77) but not in the same family. The mother harbored both gene mutations, whereas the proband and his sister had only the THRB gene mutation and a brother only the TBG gene mutation. This family illustrates the difficulty that might be encountered in the interpretation of thyroid function tests when different genetic defects, having opposite effect on thyroid function tests, coexist in the same family, and especially the same individual.

 

TRANSTHYRETIN (TTR) DEFECTS

 

Sequencing of the TTR gene, formerly known as thyroxine-binding prealbumin (TBPA) on chromosome 18 (18q11.2-q12.1), has uncovered mutations that produce variant TTR molecules with or without alterations in the binding affinity for iodothyronines (2, 78).  Only those known to affect iodothyronine binding are listed in table 2.  Some of the TTR variants are responsible for the dominantly inherited familial amyloidotic polyneuropathy (FAP), causing multiple organ failure and death in early adulthood.(78).  Because TTR has a relatively lower affinity for T4 (about 100-fold lesser than that of TBG), it plays a minor role in thyroid hormone transport in blood.  Accordingly, changes in the TTR concentration in serum and variant TTRs with reduced affinity for T4 have little effect on the concentration of serum T4 (79, 91).  Only variant TTRs with a substantially increased affinity for iodothyronines produce significant elevation in serum T4 and rT3 concentrations and account for 2% of subjects with euthyroid hyperthyroxinemia (90).

 

Table 2.  TTR Variants with Altered Affinity for T4 and Potentially an Effect on Tests of Thyroid Function in Serum

AFFINITY FOR T4

Mutant / Normal

TTR

CONCENTRATION

CODON

Number

AMINO ACID

(Normal - Variant)

REFERENCES

 

HOMO*

HETERO*

 

DECREASED

<0.1

0.17 - 0.41

N

30

Val - Met

(79, 80)

 

0.54

 

58

Leu - His

(80)

 

0.45

 

77

Ser - Tyr

(80)

 

0.19 – 0.46

N

84

Ile - Ser

(79, 80)

~1.0

0.44

 

 

Val - Ile

(80)

INCREASED

 

 

3.5†

N

6

Gly - Ser

(81-83)

8.3-9.8

3.2 - 4.1‡

N

109

Ala - Thr

(80, 84-86)

 

 

N

109

Ala - Val

(85)

 

 

Inc or N

119

Thr - Met

(87-90)

*  HOMO, homozygous; HETERO, heterozygous.

† Probably overestimated since the subjects harboring this TTR variant have normal serum TT4 concentrations.

‡  Affinity of recombinant TTR Thr109 is 9-fold that of the normal TTR (86).

Variant TTR tested and shown not to have altered affinity to T4 are: Ala60, (hetero) (79, 80).

N, normal; Inc, increased

Endonucleases useful in the identification of TTR variants: Msp I -ve for Ser6 in exon 2 associated PHA; BsoFI -ve and Fnu 4H +ve for Thr109; BsoFI -ve for Val109 and Nco I +ve for Met119, all in exon 4.

 

A family with elevated total T4 concentration which was predominantly bound to TTR was first described in 1982 by Moses et al (92).  The inheritance was autosomal dominant and affected members were clinically euthyroid with normal free T4 levels measured by equilibrium dialysis.  The variant TTR has a single nucleotide substitution replacing the normal Ala109 with a Thr which increases its affinity for T4, rT3 and tetraiodothyroacetic acid and to a lesser extend T3 and triiodothyroacetic acid (84, 86).  Crystallographic analysis of this variant TTR revealed an alteration in the size of the T4-binding pocket (93).  Another TTR gene mutation involving the same codon has been subsequently described (85).  This mutant TTR with Val109 has an increased affinity for T4 that is of similar magnitude as TTR Thr109, about 10-fold higher than that of wild-type TTR.

 

A more common defect found in subjects with prealbumin associated hyperthyroxinemia (PAH) is a point mutation in exon 4 of the TTR gene replacing the normal Thr119 with Met (90).  First described in a single individual with normal serum total and free T4 levels (89), the majority of subsequently identified heterozygous subjects harboring the TTR Met119 had an increase in the fraction of T4 and rT3 associated with TTR, but only few had serum T4 levels above the upper limit of normal.  Furthermore, their hyperthyroxinemia appears to be transient, usually in association with non-thyroidal illness  (90).  The variant TTRs associated with PAH are not amyloidogenic.

 

The unique occurrence in an Argentinian family of TTR A109T, known to have increased affinity for T4, in association with TGB-PD A64D mitigated the phenotype of the latter mutation (42)

 

Variant TTRs without Known Biological Effects

 

Several TTR variants have been found that do not alter the properties of the molecule, nor cause FAP, and are thus of no clinical significance.  Of interest is a TTR variant found in the rhesus monkey, Macaca mulatta, but not in man (94, 95)[.  This variant has a slower electrophoretic mobility resulting in three phenotypes which exhibit: (a) a single rapidly migrating band similar to that found in human and other primates (PAFF); (b) a single slowly migrating band cathodal to albumin (PASS); and (c) a five banded form corresponding to the various tetrameric recombinants present in the heterozygous state possessing the two different subunits (PAFS).  This finding was important because the variant rhesus PA-S could be hybridized in vitro with human TTR yielding a five-banded pattern hence, demonstrating that human TTR is also a tetramer.  All naturally occurring and hybrid polymorphic variants show no detectable alteration in the binding of either T4 or retinol binding protein  (96).

 

HUMAN SERUM ALBUMIN (HSA) DEFECTS

 

Another form of dominantly inherited euthyroid hyperthyroxinemia, later to be linked to the albumin gene on chromosome 4 (4q11-q13), was first described in 1979 (97, 98).  Known as familial dysalbuminemic hyperthyroxinemia (FDH) (99), it is the most common cause of inherited increase in total T4 in serum in the Caucasian population, producing on the average a 2-fold increase in the serum total T4 concentration.  In a study of 430 subjects suspected of having euthyroid hyperthyroxinemia 12% were proven to have FDH (90).  The prevalence varies from 0.01 to 1.8%, depending on the ethnic origin, with the highest prevalence in Hispanics (100-103).  This form of FDH has not been reported in subjects of African origin and the isolated occurrence in a Chinese (104) was possibly brought by Hispanic travelers (see below).  The euthyroid status of subjects with FDH has been confirmed by normal TSH response to TRH, normal free T4 concentration measured by equilibrium dialysis using appropriate buffer systems, normal T4 production rate and normal serum sex hormone-binding globulin concentration (97, 99, 105, 106).  Nevertheless, the falsely elevated free T4 values, when estimated by standard clinical laboratory techniques, has often resulted in inappropriate thyroid gland ablative or drug therapy (107-109).  A recent survey of commonly used commercial tests for measurement of free T4 indicates that equilibrium or symmetric dialysis are the only tests that will consistently provide accurate values in subjects with FDH (110), in particular one using dialysis in combination with tandem mass spectrometry (111).

 

FDH is suspected when serum total T4 concentration is increased without proportional elevation in total T3 level and non-suppressed serum TSH.  Half of affected subjects have also rT3 values above the normal range (112) (Table 3). Since the same combination of test results are found in subjects with the Thr109 TTR variant, the diagnosis of FDH should be confirmed by the demonstration that an increased proportion of the total serum T4 migrates with HSA on non-denaturing electrophoresis or precipitates with anti-HSA serum.

 

Table 3.  Albumin Variants with Increased Affinities for Iodothyronines, Their Effect on the Serum Concentrations of and Affinities to these Hormones

 

VARIANT

SERUM CONCENTRATION

 

 

BINDING AFFINITY (Ka)

of the variant albumins

Reference

T4

µg/dl

T3

ng/dl

rT3

ng/dl

N

T4

T3

 

 

(Fold the normal mean)

 

(Fold the normal mean)

WT

8.0 ± 0.2

125 ± 4

22.5 ± 0.9

83

1

1

A

R218H

16.0 ± 0.5

(2.0)

154 ± 3

(1.2)

33.1 ± 1.1

(1.5)

83

(10 – 15)

(4)

a,(113, 114)

R218P

135 ± 17

(16.8)

241 ± 25

(1.9)

136 ± 13

(6.1)

8

(11-13*)

(1.1*)

(9, 10)

R218S

70

(8.8)

159

(1.3)

55.7

(2.6)

1

NM

NM

(11)

R222I

21±1.4

(2.6)

135±18

(1.2)

1417±107

(86)

8

NM

NM

a, (115)

L66P

8.7

(1.1)

320

(3.3)

22.3

(1)

6

(1.5)

(40)

(3)

Values reported are means ± standard error, and the number of subjects per genotype are indicated under ‘‘N.’’

* Determined at saturation. Affinities are higher at the concentrations of T4 and T3 found in serum.

NM, not measured

a, Personal observation

All data were generated in the Chicago laboratory except for 4 of the 8 individuals with ALB R218P and hose with ALB R222I, provided by Nadia Schoenmakers, University of Cambridge, UK.

 

A tight linkage between FDH and the HSA gene (lod score 5.25) was found in a large Swiss-Amish family using two polymorphic markers (112).  This was followed by the identification of a missense mutation in codon 218 of the HSA gene replacing the normal arginine with a histidine (R218H) (113, 116).  Furthermore, the same mutation was present in all subjects with FDH from 11 unrelated families.  Its association with a Sac I+ polymorphism, suggest a founder effect and is compatible with ethnic predilection of FDH (113).  The coexistence of FDH and a TTR variant with increased affinity for T4 in the same individual (82, 83) and FDH with TBG-PD in another (117) have been reported.  In both instances these individuals were the product of parents each heterozygous for of one of the two defects.

 

Another mutation in codon 218, with increased affinity to iodothyronines, was first identified by Wada et al (9).  The mutation, a replacement of the normal Arg218 with a Pro (R218P), initially believed to be unique for Japanese was also identified in a Swiss family with no Asian ancestry (Fig. 5) (10).  In this form of FDH, serum total T4 levels are 14-20-fold the normal mean, a level confirmed by measurements in serum extracts by HPLC.  Total rT3 and T3 concentrations are 7- and 2-fold above the mean, respectively.  Thus, in order to maintain a normal free T4 level, the calculated affinity constant (Ka) of HSA R218P should be about 16-fold higher than that of HSA R218H.  Surprisingly, the Kas measured at saturation were similar, 5.4 x 106 M-1 and 6.4 x 106 M-1 for HSA R218H, respectively (10, 118, 119) (Table 3).  However, at T4 concentrations equivalent to those found in subjects with HSA R218P, the dialyzable FT4 concentration was 11-fold higher in serum of subjects with HSA R218H and 49-fold higher in serum with the common type HSA only (10).

 

Figure 5. A Swiss family with HSA R218P: genotyping, pedigree and thyroid function tests. A, Genotyping for the mutation HSA R218P. Results are aligned with each subject depicted on the pedigree in B. Amplification of a segment of the HSA gene containing the mutation with a mismatched oligonucleotide primer creates a new restriction site for Ava II only in the presence of the mutant nucleotide (CGC -> CCC). Affected subjects expressing proline 218 (CCC) show a 122 bp DNA fragment produced by enzymatic digestion of the mutant allele. Note that all affected subjects are heterozygous and that the 153 bp fragment amplified from DNA of the two normal subjects, expressing arginine 218 (CGC) only, resists enzymatic digestion. B, Pedigree of the family. Roman numerals indicate each generation and numbers below each symbol identify the subject. Individuals expressing the FDH phenotype are indicated by half-filled symbols. C, Thyroid function tests. Results are aligned with each symbol. Values outside the normal range are in bold numbers. Note the disproportionate increase in serum T4 concentration as compared to that of T3 in the affected individuals. Subject I-1, a year old man, had diabetes mellitus with multiple organ complications and mild subclinical hypothyroidism, explaining the relatively lower serum T4 and T3 but not rT3 levels. [Adapted from Pannain et al (10)].

 

More recently two additional HSA gene mutations have been identified.  One in the same codon resulting in a different amino acid substitution (R218S) (11) and another in a different amino acid (R222I) (115) in the proximity of the same iodothyronine-binding pocket (Fig. 6).  While both manifest increased affinity for T4 and rT3, it is considerably higher for T4 in the former and for rT3 in the latter (Table 3). It is of note that the two amino acids, 218 and 222, involved in the gain-of-function mutations are located in the main predominantly hydrophobic pocket where T4 is bound in a cisoid conformation (120).

 

Figure 6. The structures of HSA in the presence of T4 as modeled on the structures 1BM0, 1HK1, 1HK3 in the Protein Data Bank (http://www.rcsb.org/pdb/home/home.do). Top panel shows on the left the entire WT HSA molecule (in green) with its four T4 binding sites [T4(1) to T4(4)] according to Petitpas et al (120) and to the right a close up of the binding pocket, T4 (1) containing arginine’s 218 and 222 along with the T4 molecule (carbons are in white, nitrogen’s in blue, oxygens in red and iodine in magenta). In the bottom panel are represented the structures of the T4 (1) binding pockets of the four mutant HSA showing, a better accommodation of T4 than in the WT HSA and thus, resulting in enhanced binding (From Erik Schoenmakers, University of Cambridge, UK).

 

A fifth gain-of-function mutation, a replacement of the normal Leu66 with a Pro (L66P) has been identified in a single Thai family (3).  It produces a 40-fold increase in the affinity for T3 but only 1.5-fold increase in the affinity for T4 (Table 3).  As a consequence, patients have hypertriiodothyroninemia but not hyperthyroxinemia.  In this FDH-T3, serum T3 concentrations are falsely low, or even undetectable, when T3 is measured using an analog of T3 as a tracer rather than a radioisotope.  It has resulted in the inappropriate treatment with thyroid hormone (3).

 

Bisalbuminemia and Analbuminemia

 

Variant albumins, with altered electrophoretic mobility produce "bisalbuminemia" in the heterozygotes (121).  T4 binding has been studied in subjects from unrelated families with a slow HSA variant.  In two studies only the slow moving HSA bound T4 (122, 123) and in another, both (124).  The differential binding of T4 to one of the components of bisalbumin may be due to enhanced binding to the variant component with charged amino acid sequence.  Bisalbuminemia does not seem to be associated with gross alterations in thyroid hormone concentration in serum.

 

Analbuminemia is extremely rare, occurring in less than 1 in a million individuals (125).  The first case was reported in 1954 (126) but the HSA gene mutation was identified 56 years later (127).  The less than 50 cases so far reported have nonsense mutations causing premature termination of translation or splicing defects (128).  Despite the complete lack of such an important substance, symptoms are remarkably mild owing to the compensation by an increase in non-albumin serum proteins.  Studies with respect to T4-transport showed no clear effect or slight increase total serum iodothyronines, associated with increased levels of TBG and TTR. (128, 129).  The latter two normalized when serum HSA was restored to normal by multiple transfusions (129).

 

ACKNOWLEDGMENTS

 

Supported in part by grants DK-15070 from the National Institutes of Health (USA).

 

REFERENCES

 

  1. Beierwaltes WH, Robbins J 1959 Familial increase in the thyroxine-binding sites in serum alpha globulin. J Clin Invest 38:1683-1688.
  2. Pappa T, Ferrara AM, Refetoff S 2015 Inherited defects of thyroxine-binding proteins. Best Pract Res Clin Endocrinol Metab 29:735-747.
  3. Sunthornthepvarakul T, Likitmaskul S, Ngowngarmratana S, Angsusingha K, Sureerat K, Scherberg NH, Refetoff S 1998 Familial dysalbuminemic hypertriiodothyroninemia: a new dominantly inherited albumin defect. J Clin Endocrinol Metab 83:1448-1454.
  4. Refetoff S 1989 Inherited thyroxine-binding globulin (TBG) abnormalities in man. Endocr Rev 10:275-293.
  5. Refetoff S, Robin NI, Alper CA 1972 Study of four new kindreds with inherited thyroxine-binding globulin abnormalities: Possible mutations of a single gene locus. J Clin Invest 51:848-867.
  6. Burr WA, Ramsden DB, Hoffenberg R 1980 Hereditary abnormalities of thyroxine-binding globulin concentration. Quart J Med 49:295-313.
  7. Trent JM, Flink IL, Morkin E, Van TuinenP, Ledbetter  DH 1987 Localization of the human thyroxine-binding globulin gene to the long arm of the X chromosome (Xq21-22). Am J Hum Genet 41:428-435.
  8. Mori Y, Miura Y, Oiso Y, Seo H, Takazumi K 1995 Precise localization of the human thyroxine-binding globulin gene to chromosome Xq22.2 by fluorescence in situ hybridization. Hum Genet 96:481-482.
  9. Wada N, H. C, Shimizu C, Kijima H, Kubo M, Koike T 1997 A novel misssense mutation in codon 218 of the albumin gene in a distinct phenotype of familial dysalbuminemic hyperthyroxinemia in a Japanese kindred. J Clin Endocriol Metab 82:3246-3250.
  10. Pannain S, Feldman M, Eiholzer U, Weiss RE, Scherberg NH, Refetoff S 2000 Familial dysalbuminemic hyperthyroxinemia in a Swiss family caused by a mutant albumin (R218P) shows an apparent discrepancy between serum concentration and affinity for thyroxine. J Clin Endocrinol Metab 85:2786-2792.
  11. Greenberg SM, Ferrara AM, Nicholas ES, Dumitrescu AM, Cody V, Weiss RE, Refetoff S 2014 A Novel Mutation in the Albumin Gene (R218S) Causing Familial Dysalbuminemic Hyperthyroxinemia in a Family of Bangladeshi Extraction. Thyroid 24:945-950.
  12. Okamoto H, Mori Y, Tani Y, Nakagomi Y, Sano T, Ohyama K, Saito H, Oiso Y 1996 Molecular analysis of females manifesting thyroxine-binding globulin (TBG) deficiency: selective X-chromosome inactivation responsible for the difference between phenotype and genotype in TBG-deficient females. J Clin Endocrinol Metab 81:2204-2208.
  13. Gawandi S, Jothivel K, Kulkarni S 2022 Identification of a novel mutation in thyroxine-binding globulin (TBG) gene associated with TBG-deficiency and its effect on the thyroid function. J Endocrinol Invest 45:731-739.
  14. Refetoff S, Selenkow HA 1968 Familial thyroxine-binding globulin deficiency in a patient with Turner's syndrome (X0): Genetic study of a kindred. N Engl J Med 278:1081-1087.
  15. Reutrakul S, Janssen OE, Refetoff S 2001 Three novel mutations causing complete T4-binding globulin deficiency. J Clin Endocrinol Metab 86:5039-5044.
  16. Mannavola D, Vannucchi G, Fugazzola L, Cirello V, Campi I, Radetti G, Persani L, Refetoff S, Beck-Peccoz P 2006 TBG deficiency: description of two novel mutations associated with complete TBG deficiency and review of the literature. J Mol Med 84:864-871.
  17. Moeller LC, Appiagyei-Dankah Y, Kohler B, Biebermann H, Janssen OE, Fuhrer D 2015 Two Novel Mutations in the Serpina7 Gene Are Associated with Complete Deficiency of Thyroxine-Binding Globulin. Eur Thyroid J 4:108-112.
  18. Domingues R, Bugalho MJ, Garrão A, Boavida JM, Sobrinho L 2002 Two novel variants in thyroxine-binding globulin (TBG) gene behind the diagnosis of TBG deficiency. Eur J Endocrinol 146:485-490.
  19. Ueta Y, Mitani Y, Yoshida A, Taniguchi S, Mori A, Hattori K, Hisatome I, Manabe I, Takeda K, Sato R, Ahmmed GU, Tsuboi M, Ohtahara A, Hiroe K, Tanaka Y, Shigemasa C 1997 A novel mutation causing complete deficiency of thyroxine binding globulin. Clin Endocrinol (Oxf) 47:1-5.
  20. Hershkovitz E, Leiberman E, Refetoff S, Pilpell D, Phillip M 1995 High prevalence of thyroxine-binding globulin deficiency among Bedouin infants in southern Israel. Isr J Med Sci 31:500-502.
  21. Miura Y, Hershkovitz E, Inagaki A, Parvari R, Oiso Y, Phillip M 2000 A novel mutation causing complete thyroxine-binding globulin deficiency (TBG-CD-Negev) among the Bedouins in southern Israel. J Clin Endocrinol Metab 85:3687-3689.
  22. Su CC, Wu YC, Chiu CY, Won JG, Jap TS 2003 Two novel mutations in the gene encoding thyroxine-binding globulin (TBG) as a cause of complete TBG deficiency in Taiwan. Clin Endocrinol (Oxf) 58:409-414.
  23. Li P, Janssen OE, Takeda K, Bertenshaw RH, Refetoff S 1991 Complete thyroxine-binding globulin (TBG) deficiency caused by a single nucleotide deletion in the TBG gene. Metabolism 40:1231-1234.
  24. Carvalho GA, Weiss RE, Refetoff S 1998 Complete thyroxine-binding globulin (TBG) deficiency produced by a mutation in the acceptor splice site causing frameshift and early termination of translation (TBG-Kankakee). J Clin Endocrinol Metab 83:3604-3608.
  25. Lacka K, Nizankowska T, Ogrodowicz A, Lacki JK 2007 A Novel Mutation (del 1711 G) in the TBG Gene as a Cause of Complete TBG Deficiency. Thyroid 17:1143-1146.
  26. Mori Y, Takeda K, Charbonneau M, Refetoff S 1990 Replacement of Leu227 by Pro in thyroxine-binding globulin (TBG) is associated with complete TBG deficiency in three of eight families with this inherited defect. J Clin Endocrinol Metab 70:804-809.
  27. Domingues R, Font P, Sobrinho L, Bugalho MJ 2009 A novel variant in Serpina7 gene in a family with thyroxine-binding globulin deficiency. Endocrine 36:83-86.
  28. Personal-Observation.
  29. Carvalho GA, Weiss RE, Vladutiu AO, Refetoff S 1998 Complete deficiency of thyroxine-binding globulin (TBG-CD Buffalo) caused by a new nonsense mutation in the thyroxine-binding globulin gene. Thyroid 8:161-165.
  30. Reutrakul S, Dumitrescu A, Macchia P, Moll GWJ, Vierhapper H, Refetoff S 2002 Complete thyroxine-binding globulin (TBG) deficiency in two families without mutations in coding or promoter regions of the TBG gene: in vitro demonstration of exon skipping. J Clin Endocrinol Metab 87:1045-1051.
  31. Yamamori I, Mori Y, Seo H, Hirooka Y, Imamura S, Miura Y, Matsui N, Oiso Y 1991 Nucleotide deletion resulting in frameshift as a possible cause of complete thyroxine-binding globulin deficiency in six Japanese families. J Clin Endocrinol Metab 73:262-267.
  32. Yamamori I, Mori Y, Miura Y, Tani Y, Imamura S, Oiso Y, Seo H 1993 Gene screening of 23 Japanese families with complete thyroxine-binding globulin deficiency: Identification of a nucleotide deletion at codon 352 as a common cause. Endocr J 40:563-569.
  33. Yorifuji T, Muroi J, Uematsu A, Momoi T, Furusho K 1998 Identification of a novel variant of the thyroxine-binding globulin (TBG) in a Japanese patient with TBG deficiency (abstract). Horm Res 50 (suppl 3):68.
  34. Murata Y, Takamatsu J, Refetoff S 1986 Inherited abnormality of thyroxine-binding globulin with no demonstrable thyroxine-binding activity and high serum levels of denatured thyroxine-binding globulin. N Engl J Med 314:694-699.
  35. Moeller LC, Fingerhut A, Lahner H, Grasberger H, Weimer B, Happ J, Mann K, Janssen OE 2006 C-Terminal Amino Acid Alteration rather than Late Termination Causes Complete Deficiency of Thyroxine-Binding Globulin CD-NeuIsenburg. J Clin Endocrinol Metab 91:3215-3218.
  36. Fingerhut A, Reutrakul S, Knuedeler SD, Moeller LC, Greenlee C, Refetoff S, Janssen OE 2004 Partial deficiency of thyroxine-binding globulin-allentown is due to a mutation in the signal Peptide. J Clin Endocrinol Metab 89:2477-2483.
  37. Bertenshaw R, Sarne D, Tornari J, Weinberg M, Refetoff S 1992 Sequencing of the variant thyroxine-binding globulin (TBG)-San Diego reveals two nucleotide substitutions. Biochim Biophys Acta 1139:307-310.
  38. Janssen OE, Astner ST, Grasberger H, Gunn SK, Refetoff S 2000 Identification of thyroxine-binding globulin-San Diego in a family from Houston and its characterization by in vitro expression using Xenopus oocytes. J Clin Endocrinol Metab 85:368-372.
  39. Sarne DH, Refetoff S, Nelson JC, Dussault J 1989 A new inherited abnormality of thyroxine-binding globulin (TBG-San Diego) with decreased affinity for thyroxine and triiodothyronine. J Clin Endocrinol Metab 68:114-119.
  40. Gomes Lima CJ, de Olivera Andrada M, Santos daCunha V, Linhares Maciel AAF, Lofrano-Porto A 2015 A novel variant in the SERPINA7 gene is associated with TBG partial deficiency in a woman and her two male siblings ENDO 2015, San Diego CA, FRI-068.
  41. Moeller LC, Vinzelberg P, Jaeger A, Fingerhut A, Mann K, Janssen OE 2008 Two novel mutations leading to partial and complete thyroxine-binding globulin deficiency Symposium of the German Society for Endocrinology, Salzburg, Austria

.

  1. Sklate RT, Olcese MC, Maccallini GC, Sarmiento RG, Targovnik HM, Rivolta CM 2014 Novel mutation p.A64D in the Serpina7 gene as a cause of partial thyroxine-binding globulin deficiency associated with increases affinity in transthyretin by a known p.A109T mutation in the TTR gene. Horm Metab Res 46:100-108.
  2. Mori Y, Seino S, Takeda K, Flink IL, Murata Y, Bell GI, Refetoff S 1989 A mutation causing reduced biological activity and stability of thyroxine-binding globulin probably as a result of abnormal glycosylation of the molecule. Mol Endocrinol 3:575-579.
  3. Takamatsu J, Refetoff S, Charbonneau M, Dussault JH 1987 Two new inherited defects of the thyroxine-binding globulin (TBG) molecule presenting as partial TBG deficiency. J Clin Invest 79:833-840.
  4. Janssen OE, Refetoff S 1992 In-vitro expression of thyroxine-binding globulin (TBG) variants: Impaired secretion of TBGPRO-227 but not TBGPRO-113. J Biol Chem 267:13998-14004.
  5. Refetoff S, Murata Y 1985 X-chromosome-linked inheritance of the variant thyroxine-binding globulin in serum of Australian Aborigines: Its physical, chemical and biological properties. J Clin Endocrinol Metab 60:356-360.
  6. Takeda K, Mori Y, Sobieszczyk S, Seo H, Dick M, Watson F, Flink IL, Seino S, Bell GI, Refetoff S 1989 Sequence of the variant thyroxine-binding globulin of Australian Aborigines: Only one of two amino acid replacements is responsible for its altered properties. J Clin Invest 83:1344-1348.
  7. Moeller LC, Edidin DV, Jaeger A, Mann K, Refetoff S 2010 A novel mutation leading to familial partial thyroxine-binding globulin deficiency (TBG Glencoe) 14th International Thyroid Congress, Paris, France.
  8. Bertenshaw R, Takeda K, Refetoff S 1991 Sequencing of the variant thyroxine-binding globulin (TBG)-Quebec reveals two nucleotide substitutions. Am J Hum Genet 48:741-744.
  9. Miura Y, Mori Y, Yamamori I, Tani Y, Murata Y, Yoshimoto M, Kinoshita E, Matsumoto T, Oiso Y, Seo H 1993 Sequence of a variant thyroxine-binding globulin (TBG) in a family with partial TBG deficiency in Japanese (TBG-PDJ). Endocr J 40:127-132.
  10. Miura Y, Mori Y, Kambe F, Tani Y, Oiso Y, Seo H 1994 Impaired intracellular transport contributes to partial thyroxine-binding globulin (TBG) deficiency in a Japanese family. J Clin Endocrinol Metab 79:740-744.
  11. Not Published.
  12. Ferrara AM, Pappa T, Fu J, Brown CD, Peterson A, Moeller LC, Wyne K, White KP, Pluzhnikov A, Trubetskoy V, Nobrega M, Weiss RE, Dumitrescu AM, Refetoff S 2015 A novel mechanism of inherited TBG deficiency: mutation in a liver-specific enhancer. J Clin Endocrinol Metab 100:E173-181.
  13. Daiger SP, Rummel DP, Wang L, Cavalli-Sforza LL 1981 Detection of genetic variation with radioactive ligands.IV.  X-linked, polymorphic genetic variation of thyroxin-binding globulin (TBG). Am J Hum Genet 33:640-648.
  14. Takamatsu J, Ando M, Weinberg M, Refetoff S 1986 Isoelectric focusing variant thyroxine-binding globulin (TBG-S) in American Blacks: Increased heat lability and reduced concentration in serum. J Clin Endocrinol Metab 63:80-87.
  15. Waltz MR, Pullman TN, Takeda K, Sobieszczyk P, Refetoff S 1990 Molecular basis for the properties of the thyroxine-binding globulin-slow variant in American Blacks. J Endocrinol Invest 13:343-349.
  16. Takeda K, Iyota K, Mori Y, Tamura Y, Suehiro T, Kubo Y, Refetoff S, Hashimoto K 1994 Gene screening in Japanese families with complete deficiency of thyroxine-binding globulin demonstrates that a nucleotide deletion at codon 352 may be a race specific mutation. Clin Endocrinol (Oxf) 40:221-226.
  17. Janssen EO, Chen B, Büttner C, Refetoff S, Scriba PC 1995 Molecular and structural characterization of the heat-resistant thyroxine-binding globulin-Chicago. J Biol Chem 270:28234-28238.
  18. Takamatsu J, Refetoff S 1986 Inherited heat stable variant thyroxine-binding globulin (TBG-Chicago). J Clin Endocrinol Metab 63:1140-1144.
  19. Kambe F, Seo H, Mori Y, Murata Y, Janssen OE, Refetoff S, Matsui N 1992 An additional carbohydrate chain in the variant thyroxine-binding globulin-Gary (TBGAsn-96) impairs its secretion. Mol Endocrinol 6:443-449.
  20. Murata Y, Refetoff S, Sarne DH, Dick M, Watson F 1985 Variant thyroxine-binding globulin in serum of Australian Aborigines: Its physical, chemical and biological properties. J Endocrinol Invest 8:225-232.
  21. Refetoff S, Dumont JE, Vassart G 2000 Thyroid disorders. In: Scriver CR, Beaudet AL, Sly WS, Valle D, Childs B, Vogeldstein B, (eds) The Metabolic and Molecular Basis of Inherited Disease.The Metabolic and Molecular Basis of Inherited Disease. 8 ed. Vol 3. MacGraw-Hill, New York, 4029-4075.
  22. Janssen OE, Bertenshaw R, Takeda K, Weiss R, Refetoff S 1992 Molecular basis of inherited thyroxine-binding globulin defects. Trends Endocrinol Metab 3:49-53.
  23. Sarne DH, Refetoff S, Murata Y, Dick M, Watson F 1985 Variant thyroxine-binding globulin in serum of Australian Aborigines. A comparison with familial TBG deficiency in Caucasians and American Blacks. J Endocrinol Invest 8:217-224.
  24. Kobayashi H, Sakurai A, Katai M, Hashizume K 1999 Autosomally transmited low concentration of thyroxine-binding globuline. Thyroid 9:159-163.
  25. Griffiths KD, Virdi NK, Rayner PHW, Green A 1985 Neonatal screening for congenital hypothyroidism by measurement of plasma thyroxine and thyroid stimulating hormone concentrations. Brit Med J 291:117-120.
  26. Brown SK, Bellisario R 1986 Measurement of thyroxine-binding globulin (TBG) levels from dried blood spot specimens: Detection of TBG deficiency and TBG excess in infants. In: Carter TP, Wiley AH, (eds) Genetic Disease: Screening and Management. Vol. Alan R. Liss, Inc., 373-384.
  27. Refetoff S, Charbonneau M, Sarne DH, Takamatsu J, Dussault JH 1989 Resistance to thyroid hormones and screening for high thyroxine at birth. Research for Congenital Hypothyroidism. Vol. Plenum Publishing Corp., 165-172.
  28. Hayashi Y, Mori Y, Janssen OE, Sunthornthepvarakul T, Weiss RE, Takeda K, Weinberg M, Seo H, Bell GI, Refetoff S 1993 Human thyroxine-binding globulin gene: Complete sequence and transcriptional regulation. Mol Endocrinol 7:1049-1060.
  29. Mori Y, Miura Y, Takeuchi H, Igarashi Y, Sugiura J, Oiso Y 1995 Gene amplification as a cause for inherited thyroxine-binding globulin excess in two Japanese families. J Clin Endocrinol Metab 80:3758-3762.
  30. Kamboh MI, Ferrell RE 1986 A sensitive immunoblotting technique to identify thyroxine-binding globulin protein heterogeneity after isoelectric focusing. Biochem Genet 24:273-280.
  31. Constans J, Ribouchon MT, Gouaillard C, Chaventré A, Clayton J 1992 A new polymorphism of thyroxin-binding globulin in three African groups (Mali) with endemic nodular goitre. Hum Genet 89:199-203.
  32. Duhan U, Patston P 2010 Explanation for the high heat stability of thyroxine binding globulin-Chicago. Endocr Regul 44:43-47.
  33. Miura Y, Kambe F, Yamamori I, Mori Y, Tani Y, Murata Y, Oiso Y, Seo H 1994 A truncated thyroxine-binding globulin due to a frameshift mutation is retained within the rough endoplasmic reticulum: A possible mechanism of complete thyroxine-binding globulin deficiency in Japanese. J Clin Endocrinol Metab 78:283-287.
  34. Zhou A, Wei Z, Read RJ, Carrell RW 2006 Structural mechanism for the carriage and release of thyroxine in the blood. Proc Natl Acad Sci U S A 103:13321-13326.
  35. Ferrara AM, Cakir M, Henry PH, Refetoff S 2013 Coexistence of THRB and TBG Gene Mutations in a Turkish Family. J Clin Endocrinol Metab 98:E1148-1151.
  36. Adams M, Matthews C, Collingwood TN, Tone Y, Beck-Peccoz P, Chatterjee KK 1994 Genetic analysis of 29 kindreds with generalized and pituitary resistance to thyroid hormone. Identification of thirteen novel mutations in the thyroid hormone receptor beta gene. J Clin Invest 94:506-515.
  37. Benson MD 1995 Amyloidosis (Chapter131). In: Scriver CR, Beaudet AL, Sly WS, Valle D, (eds) The Metabolic and Molecular Basis of Inherited Disease. Vol. McGraw Hill Inc., New York.
  38. Refetoff S, Dwulet FE, Benson MD 1986 Reduced affinity for thyroxine in two of three structural thyroxine-binding prealbumin variants associated with familial amyloidotic polyneuropathy. J Clin Endocrinol Metab 63:1432-1437.
  39. Rosen HN, Moses AC, Murrell JR, Liepnieks JJ, Benson MD 1993 Thyroxine interactions with transthyretin: a comparison of 10 naturally occurring human transthyretin variants. J Clin Endocrinol Metab 77:370-374.
  40. Akbari MT, Fitch NJ, Farmer M, Docherty K, Sheppard MC, Ramsden DB 1990 Thyroxine-binding prealbumin gene: a population study. Clin Endocrinol (Oxf) 33:155-160.
  41. Fitch NJS, Akbary MT, Ramsden DB 1991 An inherited non-amyloidogenic transthyretin variant, [Ser6]-TTR, with increased thyroxine-binding affinity, characterized by DNA sequencing. J Endocrinol 129:309-313.
  42. Lalloz MR, Byfield PG, Goel KM, Loudon MM, Thomson JA, Himsworth RL 1987 Hyperthyroxinemia due to the coexistence of two raised affinity thyroxine-binding proteins (albumin and prealbumin) in one family. J Clin Endocrinol Metab 64:346-352.
  43. Moses C, Rosen HN, Moller DE, Tsuzaki S, Haddow JE, Lawlor J, Liepnieks JJ, Nichols WC, Benson MD 1990 A point mutation in transthyretin increases affinity for thyroxine and produces euthyroid hyperthyroxinemia. J Clin Invest 86:2025-2033.
  44. Refetoff S, Marinov VSZ, Tunca H, Byrne MM, Sunthornthepvarakul T, Weiss RE 1996 A new family with hyperthyroxinemia due to transthyretin Val109 misdiagnosed as thyrotoxicosis and resistance to thyroid hormone. J Clin Endocrinol Metab 81:3335-3340.
  45. Rosen HN, Murrell JR, Liepnieks JL, Benson MD, Cody V, Moses AC 1994 Threonine for alanine substitution at position 109 of transthyretin differentially alters human transthyretin's affinity for iodothyronines. J Clin Endocrinol Metab 134:27-34.
  46. Alves IL, Divino CM, Schussler GC, Altland K, Almeida MR, Palha JA, Coelho T, Costa PP, Saraiva MJM 1993 Thyroxine binding in a TTR met 119 kindred. J Clin Endocrinol Metab 76:484-488.
  47. Curtis AL, Scrimshaw BL, Topliss DJ, Stockigt JR, George PM, Barlow JW 1994 Thyroxine binding by human transthyretin variants: mutations at position 119, but not 54, increase thyroxine binding affinity. J Clin Endocrinol Metab 78:459-462.
  48. Harrison HH, Gordon ED, Nichols WC, Benson MD 1991 Biochemical and clinical characterization of prealbuminCHICAGO:An apparently benign variant of serum prealbumin (transthyretin) discovered with high-resolution two-dimensional electrophoresis. Am J Med Gen 39:442-452.
  49. Scrimshaw BJ, Fellowes AP, Palmer BN, Croxson MS, Stockigt JR, George PM 1992 A novel variant of transthyretin (prealbumin), Thr119 to Met, associated with increased thyroxine binding. Thyroid 2:21-26.
  50. Woeber KA, Ingbar SH 1968 The contribution of thyroxine-binding prealbumin to the binding of thyroxine in human serum, as assessed by immunoadsorption. J Clin Invest 47:1710-1721.
  51. Moses AC, Lawlor J, Haddow J, Jackson IMD 1982 Familial euthyroid hyperthyroxinemia resulting from increased thyroxine binding to thyroxine-binding prealbumin. N Engl J Med 306:966-969.
  52. Steinrauf LK, Hamilton JA, Braden BC, Murrell JR, Benson MD 1993 X-ray crystal structure of the Ala-109 -> Thr variant of human transthyretin which produces euthyroid hyperthyroxinemia. J Biol Chem 268:2425-2430.
  53. Alper CA, Robin NI, Refetoff S 1969 Genetic polymorphism of Rhesus thyroxine-binding prealbumin: Evidence for tetrameric structure in primates. Proc Natl Acad Sci U S A 63:775-781.
  54. Bernstein RS, Robbins J, Rall JE 1970 Polymorphism of monkey thyroxine-binding prealbumin (TBPA): Mode of inheritance and hybridization. Endocrinol 86:383-390.
  55. van Jaarsveld PP, Branch WT, Edelhoch H, Robbins J 1973 Polymorphism of Rhesus monkey serum prealbumin.Molecular properties and binding of thyroxine and retinol-binding protein. J Biol Chem 248:4706-44712.
  56. Henneman G, Krenning EP, Otten M, Docter R, Bos G, Visser TJ 1979 Raised total thyroxine and free thyroxine index but normal free thyroxine. A serum abnormality due to inherited increased affinity of iodothyronines for serum binding protein. Lancet 1:639-642.
  57. Lee WNP, Golden MP, Van Herle AJ, Lippe BM, Kaplan SA 1979 Inherited abnormal thyroid hormone-binding protein causing selective increase of total serum thyroxine. J Clin Endocrinol Metab 49:292-299.
  58. Ruiz M, Rajatanavin R, Young RA, Taylor C, Brown R, Braverman LE, Ingbar S 1982 Familial dysalbuminemic hyperthyroxinemia: A syndrome that can be confused with thyrotoxicosis. N Engl J Med 306:635-639.
  59. DeCosimo DR, Fang SL, Braverman LE 1987 Prevalence of familial dysalbuminemic hyperthyroxinemia in Hispanics. Ann Intern Med 107:780-781.
  60. Jensen IW, Faber J 1988 Familial dysalbuminaemic hyperthyroxinemia: a review. J Royal Soc Med 81:34-37.
  61. Sapin R, Gasser F, Chambron J 1989 Hyperthyroxinémie familiale avec dysalbuminémie:Dépistage sur 21 000 patients a l'occasion d'un bilan thyroïden. Pathol Biol (Paris) 37:785-789.
  62. Arevalo G 1991 Prevalence of familial dysalbuminemic hyperthyroxinemia in serum samples received for thyroid testing. Clin Chem 37:1430-1431.
  63. Tang KT, Yang HJ, Choo KB, Lin HD, Fang SL, Braverman LE 1999 A point mutation in the albumin gene in a Chinese patient with familial dysalbuminemic hyperthyroxinemia. Eur J Endocrinol 141:374-378.
  64. DeNayer P, Malvaux P 1982 Hyperthyroxinemia associated with high thyroxine binding to albumin in euthyroid subjects. J Endocrinol Invest 5:383-386.
  65. Mendel CM, Cavalieri RR 1984 Thyroxine distribution and metabolism in familial dysalbuminemic hyperthyroxinemia. J Clin Endocrinol Metab 59:499-504.
  66. Fleming SJ, Applegate GF, Beardwell CG 1987 Familial dysalbuminemic hyperthyroxinemia. Postgrad Med J 63:273-275.
  67. Wood DF, Zalin AM, Ratcliffe WA, Sheppard MC 1987 Elevation of free thyroxine measurement in patients with thyrotoxicosis. Quart J Med 65:863-870.
  68. Croxson MS, Palmer BN, Holdaway IM, Frengley PA, Evans MC 1985 Detection of familial dysalbuminaemic hyperthyroxinaemia. Br Med J 290:1099-1102.
  69. Cartwright D, O'Shea P, Rajanayagam O, Agostini M, Barker P, Moran C, Macchia E, Pinchera A, John R, Agha A, Ross HA, Chatterjee VK, Halsall DJ 2009 Familial dysalbuminemic hyperthyroxinemia: a persistent diagnostic challenge. Clin Chem 55:1044-1046.
  70. Refetoff S, Scherberg NH, Yuan C, Wu W, Wu Z, McPhaul MJ 2020 Free Thyroxine Concentrations in Sera of Individuals with Familial Dysalbuminemic Hyperthyroxinemia: A Comparison of Three Methods of Measurement. Thyroid 30:37-41.
  71. Weiss RE, Angkeow P, Sunthornthepvarakul T, Marcus-Bagley D, Cox N, Alper CP, Refetoff S 1995 Linkage of familial dysalbuminemic hyperthyroxinemia to the albumin gene in a large Amish family. J Clin Endocrinol Metab 80:116-121.
  72. Sunthornthepvarakul T, Angkeow P, Weiss RE, Hayashi Y, Refetoff S 1994 A identiucal missense mutation in the albumin gene produces familial disalbuminemic hyperthyroxinemia in 8 unrelated families. Biochem Biophys Res Commun 202:781-787.
  73. Petersen CE, Scottolini AG, Cody LR, Mandel M, Reimer N, Bhagavan NV 1994 A point mutation in the human serum albumin gene results in familial dysalbuminaemic hyperthyroxinaemia. J Med Genet 31:355-359.
  74. Schoenmakers N, Moran C, Campi I, Agostini M, Bacon O, Rajanayagam O, Schwabe J, Bradbury S, Barrett T, Geoghegan F, Druce M, Beck-Peccoz P, O'Toole A, Clark P, Bignell M, Lyons G, Halsall D, Gurnell M, Chatterjee K 2014 A novel albumin gene mutation (R222I) in familial dysalbuminemic hyperthyroxinemia. J Clin Endocrinol Metab 99:E1381-1386.
  75. Petersen CE, Ha C-E, Jameson DM, Bhagavan NV 1996 Mutations in a specific human serum albumin thyroxine binding site define the structural basis of familial dysalbuminemic hyperthyroxinemia. J Biol Chem 271:19110-19117.
  76. Langsteger W, Stockigt JR, Docter R, Költringer P, Lorenz O, Eber O 1994 Familial disalbuminaemic hyperthyroxinaemia and inherited partial TBG deficiency: fist report. Clin Endocrinol (Oxf) 40:751-758.
  77. Petersen CE, Ha CE, Harohalli K, Park DS, Feix JB, Isozaki O, Bhagavan NV 1999 Structural investigations of a new familial dysalbuminemic hyperthyroxinemia genotype. Clin Chem 45:1248-1254.
  78. Petersen CE, Ha CE, Harohalli K, Park DS, Bhagavan NV 2000 Familial dysalbuminemic byperthyroxinemia may result in altered warfarin pharmacokinetics. Chem Biol Interact 124:161-172.
  79. Petitpas I, Petersen CE, Ha CE, Bhattacharya AA, Zunszain PA, Ghuman J, Bhagavan NV, Curry S 2003 Structural basis of albumin-thyroxine interactions and familial dysalbuminemic hyperthyroxinemia. Proc Natl Acad Sci USA 100:6440-6445.
  80. Peters T, Jr. 1985 Serum albumin. Adv Prot Chem 37:161-245.
  81. Sarcione EJ, Aungst CW 1962 Bisalbuminemia associated with albumin thyroxine-binding defect. Clin Chim Acta 7:297-298.
  82. Tarnoky AL, Lestas AN 1964 A new type of bisalbuminemia. Clin Chim Acta 9:551-558.
  83. Andreoli M, Robbins J 1962 Serum proteins and thyroxine-protein interaction in early human fetuses. J Clin Invest 41:1070-1077.
  84. Watkins S, Madison J, Galliano M, Minchiotti L, Putnam FW 1994 A nucleotide insertion and frameshift cause analbuminemia in an Italian family. Proc Natl Acad Sci U S A 91:2275-2279.
  85. Bennhold H, Peters H, Roth E 1954 Uber einen Fall von kompletter Analbuminaemie ohne wesentliche klinische Krankheitszichen. Verh Dtsch Gesellsch Inn Med 60:630-634.
  86. Ruhoff MS, Greene MW, Peters T 2010 Location of the mutation site in the first two reported cases of analbuminemia. Clin Biochem 43:525-527.
  87. Koot BG, Houwen R, Pot DJ, Nauta J 2004 Congenital analbuminaemia: biochemical and clinical implications. A case report and literature review. Eur J Pediatr 163:664-670.
  88. Hollander CS, Bernstein G, Oppenheimer JH 1968 Abnormalities of thyroxine binding in analbuminemia. J Clin Endocrinol Metab 28:1064-1066.

 

Effect of Pregnancy on Lipid Metabolism and Lipoprotein Levels

ABSTRACT

 

Lipoprotein lipid physiology in pregnancy has important implications for the developing fetus and newborn as well as the mother. Cholesterol and essential fatty acids are essential for normal fetal development. In pregnancy, multiple physiological changes occur that contribute to the alterations in lipid profiles of healthy, gestating women. Initially, there is an anabolic phase with an increase in lipid synthesis and fat storage in preparation for the increases in fetal energy needs in late pregnancy. During the third trimester, lipid physiology transitions to a net catabolic phase with a breakdown of fat deposits. The catabolism increases substrates for the growing fetus. Overall, the changes in lipid physiology throughout the course of pregnancy allow for proper nutrients for the fetus and they reflect increasing insulin resistance in the mother. In a normal pregnancy, total cholesterol levels increase by approximately 50%, LDL-C by 30-40%, HDL-C by 25%, and triglycerides by 2- to 3-fold. Our understanding and appreciation of the full scope and implications of dyslipidemia in pregnancy on both maternal and fetal outcomes is not complete; however, it is well known that dyslipidemia in pregnancy is associated with adverse pregnancy outcomes affecting both maternal and fetal health. There are direct implications of dyslipidemia on perinatal outcomes as well as intricate relationships between dyslipidemia and other comorbid intrauterine conditions. There is also developing research indicating that the in-utero environment influences susceptibility to chronic diseases later in life, a concept known as “developmental programming.” Given all of these implications of dyslipidemia in pregnancy on maternal and fetal health, it is prudent to screen women for lipid disorders. The ideal time for this is before conception; if a woman has not been screening before pregnancy, the initial obstetrical visit is ideal. Abnormal lipids should be followed through pregnancy. The treatment of dyslipidemia in pregnancy is multifactorial, including diet, exercise, and weight management. Medical management is complicated by FDA classifications for medication risks to the fetus, however some evidence indicates there may be permissible pharmacological treatments for dyslipidemia in pregnancy. In certain instances, plasmapheresis or lipoprotein apheresis can be employed.

 

INTRODUCTION

 

Lipoprotein lipid physiology before and during pregnancy has important implications for the mother, the developing fetus, the newborn, and their future health. Cholesterol is important for normal fetal development. It is provided to the fetus via both endogenous and exogenous mechanisms. As our understanding of normal and abnormal lipid metabolism in pregnancy improves, it is clear that abnormal lipid metabolism reflected as dyslipidemia is associated with adverse perinatal outcomes. Dyslipidemia has profound associations with other pathologies in pregnancy, most notably hypertensive disorders and gestational diabetes. There is accumulating evidence of the impact of hyperlipidemia in pregnancy on the epigenetic programming of a fetus and the subsequent risk for atherogenesis for the mother and her offspring.

 

CHOLESTEROL AND OTHER LIPIDS IN FETAL DEVELOPMENT

 

Cholesterol plays a key role in the formation of cell membranes. It is essential for the formation of cell membranes, maintaining membrane integrity, and preserving cholesterol-rich domains essential for most membrane-associated signaling cascades, including sonic hedgehog signaling (1). It is also the precursor for many important hormones, such as steroids, vitamin D, and bile acids. 

           

There are multiple sources of fetal cholesterol. A significant portion is produced de novo by the fetus. Defects affecting cholesterol biosynthesis are associated with many, sometimes lethal, birth defects (2,3). Both endogenous and exogenous sources are important to fetal cholesterol homeostasis, as illustrated by a number of lines of evidence.  Cholesterol in the maternal circulation, which similarly has endogenous and exogenous sources, contributes significantly to the fetal cholesterol pool in animals and in humans (4,5). Interestingly, Vuorio and colleagues noted that concentrations of plant stanols in the cord blood of healthy newborns were 40% to 50% lower than the maternal levels (6). Because the plant stanols evaluated can only be derived from the maternal diet, placental transfer is illustrated. Fetuses with null-null mutations of 7-dehydrocholesterol reductase (Smith-Lemli-Opitz syndrome), a disorder characterized by an inability to synthesize endogenous cholesterol at normal rates, have measurable amounts of cholesterol in their bodies. This also illustrates maternal derivation (7,8). The umbilical vein, which carries blood to the fetus, has higher levels of LDL-C than the umbilical artery (9).

 

For exogenous cholesterol to be available for fetal use, it must be transported across the tissues separating the mother and fetus. Early in pregnancy, the yolk sac is the site of the transport system between the two (10). Approaching 8 weeks of gestation, the placenta becomes fully functional and takes over as the nutrient transporter (10). The transfer of lipids across the placenta and yolk sac under normal and abnormal circumstances is complex and is still incompletely understood. Cholesterol is taken up on trophoblasts’ apical or maternal side via receptor-mediated and receptor-independent transport processes. Apolipoprotein lipids are then transported across cellular barriers and delivered into the fetal circulation on the basolateral, or fetal, side of trophoblasts (4,10,11). Cultured trophoblast cells express low-density lipoprotein (LDL) receptors (LDLRs), and LDLR-cholesterol taken up by endothelial cells is well understood. How placental endothelial cells transport and deliver substantial amounts of cholesterol to the fetal microcirculation and regulate the efflux of cholesterol is undergoing intense study.

 

As opposed to adults, high-density lipoprotein (HDL) is the main cholesterol-carrying lipoprotein in fetal circulation. It differs from adult HDL by its higher proportion of apolipoprotein (Apo) E (12), but lower proportion of Apo A1 (13). The major HDL receptor, scavenger receptor class B type I (SR-BI), contributes to local cholesterol homeostasis. Arterial endothelial cells (ECA) from the human placenta are enriched with cholesterol compared to venous endothelial cells (ECV). Moreover, umbilical venous and arterial plasma cholesterol levels differ markedly. There is elevated SR-BI expression and protein abundance in endothelial cell arteries compared to veins in situ and in vitro. Immunohistochemistry demonstrated that SR-BI is mainly expressed on the apical side of placental endothelial cells in situ, allowing interaction with mature HDL circulating in the fetal blood (14). This was functionally linked to a higher increase of selective cholesterol ester uptake from fetal HDL in endothelial arteries than in endothelial veins and resulted in increased cholesterol availability in ECA. SR-BI expression on endothelial veins tended to decrease with shear stress, which, together with heterogeneous immunostaining, suggests that SR-BI expression is locally regulated in the placental vasculature (15).

 

Changes in maternal vasculature enable an increased uterine blood flow, placental nutrient, and oxygen exchange, and subsequent fetal development. Potassium (K+) channels seem to be important modulators of vascular function, promoting vasodilation, inducing cell proliferation, and regulating cell signaling (16). Different types of K(+) channels, such as Ca(2+)-activated, ATP-sensitive, and voltage-gated, have been implicated in the adaptation of maternal vasculature during pregnancy. Conversely, K(+) channel dysfunction has been associated with vascular-related complications of pregnancy, including intrauterine growth restriction and pre-eclampsia. It is thought that vascular ischemia may lead to inflammation important in pregnancy complications. Abnormalities in these pregnancy-associated vascular dilation and remodeling processes are associated with the pregnancy complications of intrauterine growth restriction (IUGR) and pre-eclampsia, in which, the normal vasodilatory effects of acetylcholine (ACh), bradykinin, Nitric oxide (NO), endothelium-derived hyperpolarizing factor (EDHF), and thromboxane-mediated responses are impaired (16).

 

Placental lipid metabolism may influence pregnancy outcomes, fetal growth, development, and life-long health (17). The placenta converts circulating maternal lipids to free fatty acids (FFAs) for uptake and processing by trophoblast cells, for metabolic demands, to produce hormones for pregnancy maintenance, and to transfer them to the developing fetus. Robust lipid uptake and metabolism early in gestation are vital to meeting the high energetic demands needed to simultaneously grow the placenta and develop embryonic organ systems. Late in gestation the human fetus requires lipids for neurodevelopment and growth, so as pregnancy progresses, metabolic adaptations in the mother and placenta uniquely support increasing lipid transport and biomagnification of essential long-chain polyunsaturated fatty acids in the last trimester (18). These fatty acids serve as local mediators of metabolism, inflammation, immune function, platelet aggregation, signal transduction, neurotransmission, and neurogenesis for the developing fetal brain and retina (19). Because these fatty acids cannot be synthesized de novo, the fetus relies on increasing placental transport, especially during the last trimester when the peak in utero accretion can surpass maternal intake to support rapid fetal brain growth. Placental lipid uptake and metabolism is a critical, highly-regulated, and surprisingly dynamic process as gestation proceeds.

 

Lipids are crucial structural and bioactive components that sustain embryo, fetal, and placental development, and growth. Intrauterine development can be disturbed by several diseases that impair maternal lipid homeostasis and lead to abnormal lipid concentrations in fetal circulation. Deficiency in essential fatty acids can lead to congenital malformations and visual and cognitive problems in the newborn. Either deficient mother-to-fetus lipid transfer or abnormal maternal-fetal lipid metabolism can cause fetal growth restriction. On the other hand, excessive mother-to-fetus fatty acid transfer can induce fetal overgrowth and lipid overaccumulation in different fetal organs and tissues. The placenta plays a fundamental role in the transfer of lipid moieties to the fetal compartment and is affected by maternal diseases associated with impaired lipid homeostasis. Studies investigating the relationship between gestational dyslipidemia and small for gestational age (SGA) have reported differing results. A recent meta-analysis found that gestations complicated with lower concentrations of TC, TG, and LDL-C, were at significantly higher risk of delivery of SGA (20). Postnatal consequences may be evident in the neonatal period or later in life. Indeed, both defects and excess of different lipid species can lead to the intrauterine programming of metabolic and cardiovascular diseases in the offspring (21)

 

NORMAL LIPID CHANGES IN A PREGNANT MOTHER

 

Figure 1 shows the average values of total cholesterol, triglycerides, low-density lipoprotein cholesterol (LDL-C), and high-density lipoprotein cholesterol (HDL-C) measured in normal women from pre-conception through several months postpartum. These values were from measurements in a cohort of women proceeding through normal pregnancy and delivery in the U.S. Circulating levels may differ depending upon the nutritional environment. It is reported that most lipoprotein concentrations increase throughout pregnancy in Gambian women for example yet are lower vs. U.S. women, the exception being medium-sized LDL and HDL particle concentrations which decrease during gestation and are similar in both cohorts (22). In a careful evaluation of normal pregnant women in Oklahoma traversing through pregnancy and delivering normal viable infants, we found that non-HDL particles, very small highly atherogenic LDL particles (small dense LDL), and total and active pcsk9 levels increase as pregnancies progress (23). BMI, an indicator of obesity, was associated with higher levels of atherogenic lipoproteins during each trimester.  Most of the women in the Wiznitzer and Wild cohorts were of young reproductive age at the time of sampling and thus their values overlap values before pregnancy and are considered in the normal range for a nonpregnant woman. In the first trimester, there is a discernible decrease in levels during the first 6 weeks of pregnancy. As pregnancy progresses, there is a noticeable increase by the third month or at the end of the first trimester. There is a steady increase throughout pregnancy. By the third trimester or near the end of pregnancy (term), levels peak (24,25). Levels of lipoprotein particles and lipids, particularly in the later part of pregnancy, are in the atherogenic range when compared to non-pregnant levels in women of comparable ages without medical conditions. After delivery, lipid and lipoprotein levels rapidly return to normal. The changes in lipid metabolism throughout pregnancy allow for proper nutrients for the fetus and the normal, steady increase throughout pregnancy is associated with increased insulin resistance in the mother. Regardless of dietary differences in cholesterol, by late pregnancy, plasma cholesterol levels are approximately 50% higher than routinely seen pre-pregnancy while triglyceride levels are increased 2-3 times (see figure 1 and table 1) (25). These changes can be viewed as important for the enhanced availability of substrates for the fetus (26,27). However, derangements in lipids are associated with adverse pregnancy outcomes and likely are associated with residual vascular damage after some key obstetrical adverse events which may put the mother at risk for later cardiovascular disease.

Figure 1. Lipid and Lipoprotein Levels During Pregnancy (Adapted from Wiznitzer A, Mayer A, Novack V, et al. Association of lipid levels during gestation with preeclampsia and gestational diabetes mellitus: a population-based study. Am J Obstet Gynecol 2009; 201(5):482.e1–8;)

 

For understanding clinical management including healthy targets, maximum plasma cholesterol values usually do not exceed 250 mg/dL during normal pregnancy, even with the marked increases in triglyceride levels that occur normally as pregnancy progresses. If abnormal pregnancies are included in cross-sectional evaluations, cholesterol levels are commonly 300 mg/dL or higher. Higher levels are consistent with a variety of maternal adverse pregnancy conditions. In normal pregnant women, the atherogenic index, LDL/HDL, remains essentially unchanged during pregnancy. This suggests that while the total lipoprotein levels increase, the cholesterol-containing lipoprotein fractions are evenly distributed (28). Physiological hyperlipidemia/hypertriglyceridemia is distinguished from pathological dyslipidemias by a paralleled increase in HDL-C in normal women as they progress through pregnancy. During pregnancy LDL and HDL are enriched in triglycerides (29). There is an increase in large HDL late in gestation and a decrease in medium HDL (30).

 

Table 1. Increase in Lipid, Lipoprotein, and Apolipoprotein Levels (from (25))

Triglycerides

2.7-fold increase

Total Cholesterol

43% increase

LDL Cholesterol

36% increase

HDL Cholesterol

25% increase

Lipoprotein (a)

190%*

Apolipoprotein B

56% increase

Apolipoprotein AI

32% increase

All increases are from 3rd trimester except the increase in HDL cholesterol

*from reference (31).

 

Small dense LDL levels increase during pregnancy, particularly in individuals who have a large increase in triglyceride levels (32,33). As one would expect given the increase in triglycerides, LDL-C, and HDL-C, apolipoprotein B, and A-I levels are also increased (table 1). In most cross-sectional studies Lp(a) levels are not elevated in pregnancy (33-35). However, in some studies that measured Lp(a) serially throughout pregnancy an increase in Lp(a) levels was observed near term (table 1) (31,36,37) (23). The failure of cross-sectional studies to find a difference in Lp(a) levels is likely due to the wide variation in Lp(a) levels between individuals.  Values in individuals range from 1mg/dL to over 200mg/dL and are largely determined by genetic factors.   

 

MECHANISMS ACCOUNTING FOR THE CHANGES IN LIPIDS DURING PREGNANCY

 

Multiple physiological changes occur during pregnancy (27,38). Hormonal and metabolic changes that occur in the mother contribute to changes in the lipid profile in healthy, gestating women. It is useful to think of two phases of lipid metabolism in normal pregnancy. During the first two trimesters, lipid metabolism is 'primarily anabolic. There is an increase in lipid synthesis and fat storage in preparation for the exponential increases in fetal energy needs in late pregnancy. This increase in lipid synthesis between 10 and 30 weeks of pregnancy is promoted by maternal hyperphagia in early pregnancy as well as an increase in insulin sensitivity. The increase in insulin sensitivity stimulates fatty acid synthesis in adipocytes and stimulates the expression of lipoprotein lipase, which results in increased uptake of fatty acids from circulating triglyceride-rich lipoproteins. Additionally, the increased production of progesterone, cortisol, leptin, and prolactin contributes to increased fat storage (24,26,27,38). There is also significant hypertrophy of the adipocytes to accommodate increased fat storage (26,27,38). 

 

Lipid metabolism in the third trimester is in a ‘net catabolic phase’, associated with a decrease in insulin sensitivity (i.e., insulin resistance) (27,38). This decrease in insulin sensitivity is associated with enhanced lipolysis of stored triglycerides in adipocytes. The third-trimester elevation of human placental lactogen (HPL) also stimulates lipolysis in adipocytes. In addition, insulin resistance results in a decrease in lipoprotein lipase in adipocytes leading to a decrease in the uptake of fatty acids from plasma triglyceride-rich lipoproteins. These changes result in a reduction in fat stored in adipocytes.

 

The hypertriglyceridemia during pregnancy is due to both the increased production and the decreased clearance of triglyceride-rich lipoproteins (27). The increased production of triglyceride-rich lipoproteins by the liver is due to the increased lipolysis of triglycerides that occurs in adipocytes, which increases free fatty acids transported to the liver. These free fatty acids are then packaged into VLDL and secreted by the liver. The high estrogen levels in the third trimester stimulate liver lipogenesis and VLDL production. Insulin resistance may also play a role in the increase in fatty acid synthesis in the liver as inhibition of glucose production can be resistant to insulin while lipogenesis is not. The increase in insulin levels that occur can stimulate hepatic fatty acid synthesis as shown in a mouse model (39).The decrease in clearance of triglyceride-rich lipoproteins is due to a decrease in lipoprotein lipase and hepatic lipase (29). The decrease in hepatic lipase is due to elevated estrogen levels (40). The decrease in lipoprotein lipase is believed to be due to a combination of factors including insulin resistance and elevated estrogen levels. The triglyceride enrichment of LDL and HDL is due to an increase in CETP activity (29) resulting in the transfer of triglyceride from VLDL to LDL and HDL and a decrease in hepatic lipase, which decreases the removal of triglycerides from these lipoprotein particles.

 

At term, LPL activity increases in the mammary glands, which will enhance the uptake of fatty acids to increase the formation of triglycerides for lactation (26).  The increase in plasma cholesterol levels is likely due to increased hepatic cholesterol synthesis (41,42). The increase in PCSK9 during pregnancy suggests an additional mechanism. The PCSK9 could result in a decrease in hepatic LDL receptors leading to increased LDL-C levels (23).

 

Table 2. Role of Hormones in Inducing Hyperlipidemia in the Third Trimester

Estrogen increase

Inhibits Hepatic Lipase

 

Stimulates VLDL production

 

Stimulates lipogenesis in the liver

Human Placental Lactogen increase

Induces insulin resistance

 

Increases lipolysis

Insulin Resistance

Decreases LPL activity

 

Increases lipolysis

 

Increase CETP

 

Stimulates lipogenesis in the liver

 

IMPLICATIONS FOR THE FETUS AND MOTHER

 

Our understanding and appreciations of the full scope and implications of dyslipidemia in pregnancy on both maternal and fetal outcomes are not complete. Maternal dyslipidemia, particularly, high triglyceride and low HDL-C levels, are associated with several adverse perinatal outcomes. To demonstrate that lipid abnormalities are causative, intervention studies to lower lipid levels demonstrating a reduction in adverse perinatal outcomes are needed. This is difficult because the study of pregnant women is protected, underfunded, and understudied, in part because of medical legal concerns and the first rule of do no harm.

 

Dyslipidemia, while asymptomatic, is an integral factor in the metabolic syndrome (MetS). Having the MetS has clear implications for maternal vascular health, and this portends a multitude of other health concerns for the mom and her fetus.

 

Gestational Diabetes

 

Pregnancy is an insulin resistance state and gestational diabetes (GDM) is thought to be unmasked due to the stress test of insulin resistance of pregnancy. Risks to the fetus from GDM include brachial plexus injuries, hypoglycemia, respiratory distress, hyperbilirubinemia, and cardiomyopathy. Women with GDM are at increased risk of pre-eclampsia and after pregnancy, a very high risk of developing overt diabetes.

 

A meta-analysis of thirteen cohorts and three nested case‐control studies found that high triglycerides early in pregnancy were associated with an increased risk of the development of GDM (43). Similarly, increasing triglycerides during pregnancy was also associated with an increased risk of developing GDM (43). An HDL‐C of <51mg/dL was associated with higher odds of GDM (43). Another meta-analysis similarly found a link between GDM and high triglyceride levels and low HDL-C levels (44). Total cholesterol levels and LDL-C levels during pregnancy were not associated with an increased risk of GDM (44).

 

In women with a history of GDM, triglyceride, total cholesterol, and LDL-C levels are increased and HDL-C levels are decreased signaling the need to closely follow lipid levels in women with a history of GDM (45). Additionally, women with GDM have an increased risk of developing cardiovascular events later in life even in the absence of developing diabetes (46)

 

Several high-risk groups may have derangements in lipid levels that put them at risk before pregnancy. In the PPCOS II study, conducted by the Reproductive Medicine Network, having the metabolic syndrome before ovulation induction for fertility enhancement was associated with a lower rate of live birth success, independent of obesity, and it was also a risk factor for pregnancy complications, in particular gestational diabetes and macrosomia (47). Efforts to reduce weight before fertility treatments have been challenging. In an obese population with unexplained infertility, although weight loss was not associated with an improvement in healthy live birth success, it was associated with a reduction in the risk of preeclampsia (48)

 

High Birth Weight

 

A review of 46 publications with 31,402 pregnancies reported that maternal high triglycerides and low HDL-C levels during pregnancy were associated with increased birthweight, a higher risk of large-for-gestational-age, macrosomia, and a lower risk of small-for-gestational-age (49). Another meta-analysis also found a link between high triglycerides and low HDL-C levels and large birthweight (50). Elevations in triglyceride levels in the first trimester are associated with increased birth weight (51). The concentration of triglycerides in the third trimester is a stronger predictor of birth weight than glucose parameters (52-54). Additionally, in women with a normal glucose tolerance test during pregnancy triglyceride levels are still predictive of birthweight (55). Elevated levels of maternal triglycerides predict macrosomia independently of other maternal factors, such as BMI and glucose levels (52-54). In contrast, total cholesterol and LDL-C levels were not predictive of large birthweight (50). Some studies suggest that high levels of maternal HDL-C are significantly associated with a decreased risk for macrosomia, perhaps indicating that HDL might have protective qualities (56,57).

 

Pre-Eclampsia

 

Pre-eclampsia is a rapidly progressive condition that affects 5-8% of pregnancies and is characterized by hypertension and proteinuria. Risks to the fetus with preeclampsia include poor fetal growth and sometimes devastating consequences of preterm birth, whether spontaneous or induced. These can manifest as cerebral palsy, epilepsy, small size, and even death.  

 

In a meta-analysis of 74 studies,  pre-eclampsia was associated with elevated total cholesterol, non-HDL-C, and triglyceride levels, regardless of gestational age at the time of blood sampling, and with lower levels of HDL-C in the third trimester (58). Other meta-analyses have confirmed the linkage of elevated triglyceride levels with pre-eclampsia (59,60).  LDL-C levels were not associated with pre-eclampsia in one meta-analysis (58) but in another meta-analysis, it was (60). Enquobahrie et. al followed a cohort of women from early pregnancy onward and found that women who developed pre-eclampsia had significantly higher concentrations of LDL-C and triglyceride levels as early as 13 weeks of gestation compared to women who remained normotensive (61). They also found that HDL-C was 7.0% lower in pre-eclamptic women than in the control group. They noted a 3.6-fold increase in risk for pre-eclampsia in women with total cholesterol >205 mg/dL, compared to women whose total cholesterol levels were <172 mg/dL, even after adjusting for confounders. Not all recent studies have found an increase in LDL-cholesterol concentration with preeclampsia during pregnancy however (62). In a secondary analysis of the FIT-PLESE randomized controlled trial, we found that elevated highly atherogenic very small LDL particles were elevated in those persons who developed pre-eclampsia after ovulation induction (63).

 

In a meta-analysis comparing women with a past history of eclampsia/pre-eclampsia vs. without, there was an increase in total cholesterol (Mean Difference = 4.6 mg/dL, CI 1.5 to 7.7), LDL-C (MD = 4.6 mg/dL; 95%CI 0.2 to 8.9), and triglycerides (MD = 7.7 mg/dL, 95%CI 3.6 to 11.7) and a decrease in HDL-C (MD = -2.15 mg/dL, 95%CI -3.46 to -0.85) (64).  It is now well recognized that women who have a history of having had eclampsia/pre-eclampsia have approximately twice the risk of cardiovascular disease later in life (65,66).

 

Preterm Birth

 

In a meta-analysis of three nested case-control studies and eight cohort studies of 13,025 pregnant women, women with elevated lipid levels were at increased risk of preterm birth (OR 1.68; 95% CI 1.25-2.26) (67). The increased risk was seen for elevated levels of total cholesterol (OR 1.71), triglycerides (OR 1.55), LDL-C (OR 1.19 not significant), and lower levels of HDL (OR 1.33). A study by Vrijkotte et al that was not included in the meta-analysis also found that elevated triglycerides but not elevated total cholesterol were associated with an increased risk of preterm birth (51).

 

Developmental Programming

 

Preclinical models demonstrate that interventions that reduce maternal cholesterol during pregnancy, that decrease oxidative stress associated with gestational dyslipidemia, or that enhance active immune defenses against oxidative stress in offspring protect against developmental programming (68-70). This is thought to be because of excess maternal cholesterol during pregnancy, all factors collectively provide evidence for causality. Early atherogenic processes in the human aorta begin during fetal development and are accelerated by dyslipidemia during pregnancy.Maternal hypercholesterolemia is associated with greatly accelerated atherogenesis in normocholesterolemic children, as shown by the FELIC study (71). In experimental models lacking the genetic and dietary variability of humans, postnatal atherosclerosis increases in proportion to the maternal cholesterol levels well into adult ages (69,72). A molecular mechanism explaining the transfer of maternal cholesterol to the fetus has been elucidated (73) and involvement of increased oxidative stress has been established (68-70).

 

The absence of routine cholesterol determinations during gestation in most countries has limited investigations of the impact of elevated maternal cholesterol during pregnancy on the clinical

manifestations of dyslipidemia in adult offspring. In the Framingham Heart Study gestational dyslipidemia in mothers was predictive of dyslipidemia in their offspring (74). Adults who had been exposed to elevated maternal LDL-C levels had 3.8 times higher odds of having elevated LDL-C levels. They found that this explained 13% of the variation in adult offspring LDL-C levels beyond common genetic variants and classic risk factors for elevated LDL-C levels. A positive association has also been reported between maternal cholesterol and newborn HDL cholesterol and subclasses (75).

 

From studies of 78 fetal aortas, maternal cholesterol explained 61% of the variance of early lesion sizes by multivariate analysis, independent of HDL-C, triglycerides, glucose, and body mass index (BMI). Maternal total cholesterol and LDLC levels were positively associated with methylation of SREBP2 in fetal aortas, suggesting a role of maternal cholesterol levels during pregnancy on epigenetic signature in offspring as reported by Napoli et al (71). SREBP2 methylation has been mapped (71). The long-term effects of maternal dyslipidemia on the progression of atherosclerosis and, its clinical manifestations are understudied. In several cohort studies, whole blood DNA methylation signatures of diet were associated with cardiovascular disease (76). Dyslipidemia can persist well into adult age and affect clinically relevant outcomes (72). In studies where elevated maternal cholesterol during pregnancy is associated with atherogenesis in childhood, maternal lipid levels during pregnancy have been associated with adult BMI, atherosclerosis-related risk, and the severity of anterior myocardial infarctions as reported by Cacciatore et al (72).

 

Pre-eclampsia (77) and gestational diabetes (46) are linked to having a greater risk for early maternal cardiovascular events.

 

LIPID SCREENING

 

The National Lipid Association (NLA) supports checking lipids routinely if there is no normal current pre-pregnancy lipid profile (78). Screening for reproductive-aged women, in general, remains deficient in part due to disparities in health services (79). Identifying pregnant women with prior atherosclerotic cardiovascular disease (ASCVD), familial hypercholesterolemia, or hypertriglyceridemia is important to allow for multi-disciplinary collaborative care. Increased knowledge and awareness are needed at both the patient and provider levels. In a survey study of 200 pregnant women within the University of Pennsylvania Health System, 59% self-reported previous lipid screening; non- Hispanic Black women were less likely to report screening (43% vs. 67%) and they had lower awareness of high cholesterol as a risk factor for ASCVD (66% vs 92%) (80). The perinatal period, when a woman sees a physician most regularly, is an opportunity to screen for lipid disorders and facilitate prevention by bringing lipid values to normal age specific target ranges. Hypertensive disorders of pregnancy are among the leading causes of maternal morbidity and mortality in the US. Pre-eclampsia, which includes hypertension and proteinuria during pregnancy, is thought to result from placental ischemia. Risk factors for pre-eclampsia parallel those for cardiovascular disease and recent studies point to hyperlipidemia, specifically hypertriglyceridemia. Current practice does not routinely include lipid testing pre-conception or during pregnancy. Professional and societal recommendations should advocate for hyperlipidemia screening, followed by appropriate management, pre-conception, and during pregnancy as an important evaluation for risk of preeclampsia during pregnancy (48).

 

A recent review recommended measuring lipids at the first visit and if normal at the beginning of third trimester (81). High risk patients should have lipids measured at first visit, beginning of second trimester, and monthly during the 3rd trimester. If triglyceride levels are greater than 250mg/dl at any time the lipid panel should be measured monthly. When and if more specialized lipid testing such as NMR or ion mobility to measure small dense LDL levels, apo B and apo A1 levels, and Lp(a) levels is needed is not defined and further studies are required. Measuring Lp(a) levels at the first visit is reasonable in patients who have not had their Lp(a) levels determined previously. If lipid abnormalities are noted during pregnancy follow-up lipid panels post-pregnancy should be obtained.

 

TREATMENT OF DYSLIPIDEMIA DURING PREGNANCY

 

Lifestyle Modifications

 

Addressing lifestyle modifications is vital in the management of any lipid disorder regardless of pregnancy status. Counseling patients regarding a heart-healthy dietary pattern that includes vegetables, fruits, whole grains, legumes, healthy protein sources, and limiting intake of sweets, sweetened beverages, and red meats along with an emphasis on weight management and exercise is essential (82). To counsel patients with elevated lipids to lower their intake of saturated fats and increase dietary fiber is important. Approaches to Stop Hypertension (DASH) diet or Mediterranean diet are well described diets that are beneficial in reducing cardiovascular risk (83). Given that women report increased motivation to enact dietary changes during pregnancy, this is an ideal time to intervene with lifestyle modifications. Diet is a critical pillar of management for hypertriglyceridemia. A very low-fat diet is recommended to mitigate the risk of pancreatitis particularly when triglyceride levels are >500 mg/dL. Severe gestational hypertriglyceridemia can lead to acute pancreatitis and the maternal mortality rate is approximately 20%.

 

Dietary interventions have benefits beyond LDL lowering due to their effects on the placenta. A low-cholesterol low-saturated fat diet in a trial of 290 pregnant patients led to a decrease in the umbilical artery pulsatility index, a method for fetal surveillance in high-risk pregnancies (less vascular resistance) (84). Increased vascular resistance is associated with adverse pregnancy outcomes such as preeclampsia, preterm delivery, and small for gestational age infants.

 

Drug Therapy

 

STATINS

 

While statins are the first-line treatment of hypercholesterolemia in the general population, their use is not recommended during pregnancy in several guidelines. The 2018 AHA/ACC/multi-society cholesterol guidelines give a class 1 recommendation that women of childbearing age with hypercholesterolemia who plan to become pregnant should stop statins 1 to 2 months before pregnancy is attempted, or if they become pregnant while on a statin, should have the statin stopped as soon as possible (85). Similarly, the European Society of Cardiology (ESC) 2019 guidelines have a class III recommendation that statin therapy is not recommended in premenopausal patients with diabetes who are considering pregnancy or are not using adequate contraception (86). Historically, statins became contraindicated in pregnancy as a result of a case series in 2004 that demonstrated an association between first-trimester statin exposure and fetal malformations. Other cohort studies of statin exposure in pregnancy have not shown an increase in teratogenic risk (87). Of all the statins, hydrophilic statins, such as pravastatin, have not been associated with anomalies (88). Meta-analyses of studies of pregnant women exposed to statins showed no increased risk of birth defects (RR 1.15) but did reveal an increased risk of miscarriage (RR 1.35) (87,89). This increased risk of miscarriage may be due to confounders such as older age and ASCVD risk factors.

 

In a retrospective review of 39 pregnancies including 20 patients with FH and 18 patients on statins, miscarriage rates were not higher in statin-exposed patients as compared to the healthy population; there was also no difference in birth weights between statin-exposed and not-exposed (90). Given the lack of clear evidence on the teratogenicity of statins, in July 2021 the FDA requested the removal of the strongest recommendation against using statins during pregnancy. They continue to advise against the use of statins in pregnancy given the limited data and quality of information of studies. The decision of whether to continue a statin during pregnancy requires shared decision-making between the patient and clinician, and healthcare professionals need to discuss the risks versus the benefits in high-risk women, such as those with homozygous FH or prior ASCVD events, that may benefit from statin therapy (91).

Discontinuation of statins in patients with FH allows cholesterol levels to increase even beyond pre-treatment levels due to higher physiologic levels during pregnancy. This period is a vulnerable time as interruptions of treatment can increase the lifelong risk of ASCVD. In women with familial hypercholesterolemia (FH), the percent increase in LDL-C levels during pregnancy is similar to that observed in women with a normal lipid profile before pregnancy even though the baseline LDL-C levels are much higher in women with familial hypercholesterolemia (92). Despite the markedly higher LDL-C levels in women with FH, the incidence of prematurity, low birth weight, and congenital malformations did not differ, however, maternal hypertension incidence was higher than for women without dyslipidemia before pregnancy who became pregnant (92,93).

 

In a retrospective review of women with homozygous familial hypercholesterolemia of which 18/39 were exposed to statins with or without ezetimibe,1 was treated with a statin, ezetimibe, and a PCSK9 inhibitor, and 5 patients were exposed to cholestyramine, and only 14 patients were not exposed to lipid-lowering therapy, complications associated with pregnancy included  3 premature infants, one preeclampsia related, the other two because of chorioamnionitis and maternal cardiac disease (90). One Intrauterine death was because of intrauterine infection.

 

More studies examining outcomes in pregnant women with FH are needed to assess short-term and long-term outcomes on the mother and the offspring. The FDA has taken a wait and see approach. Certain statins, such as pravastatin, are being investigated for use in the prevention of preeclampsia. Given the impact of statins on endothelial dysfunction and their potential ability to mediate pathways of inflammation and oxidative stress, statins are promising agents for preeclampsia treatment in persons at high risk for a severe disease which centers on vascular dysfunction. Pravastatin is the most hydrophilic statin with a short half-life and is also a substrate for multiple efflux transporters, which leads to lower transplacental transfer. The safety of pravastatin 10 mg (low-intensity) was evaluated in a very small 20-patient randomized controlled trial for the prevention of preeclampsia in high-risk pregnant women, in which the primary outcome was maternal-fetal safety. There were no differences between groups in rates of drug side effects, congenital anomalies, or other adverse events. In a larger trial, 1120 women at high risk of pre-eclampsia were randomized to either pravastatin 20 mg (low-intensity) or placebo. There was no significant reduction in the incidence of preeclampsia or differences in other biomarkers such as soluble fms-like tyrosine-kinase-1 in either treatment arm (94). Hirsch et all reviewed cohort studies assessing the effects of pravastatin on placental insufficiency disorders and found that pravastatin treatment prolonged pregnancy duration and improved associated obstetrical outcomes in pregnancies complicated with uteroplacental insufficiency disorders in cohort studies (95). Additional studies will help ascertain the efficacy of statins, such as pravastatin, during pregnancy for preeclampsia prevention.

 

BILE ACID SEQUESTRANTS

 

Bile acid sequestrants such as cholestyramine and colestipol can be used for LDL-C lowering during pregnancy (96). Since bile acid sequestrants are not absorbed they do not pass into the systemic circulation and are safer than other lipid-lowering agents. However, they do decrease the absorption of fat-soluble vitamins. Patients should be monitored for vitamin D and K deficiency (96).  According to the 2011 NLA recommendations, Colesevelam was classified as a Class B pregnancy category medication, as there are no adequate studies in pregnant women and animal studies have failed to demonstrate a risk to the fetus (97). Thus, bile acid sequestrants are considered safe for use in treating LDL-C elevations during pregnancy and breastfeeding. However, there is a lack of controlled trial data during pregnancy. Additionally, bile acid sequestrants are well known to increase triglyceride levels. They can be associated with constipation in pregnancy.

 

EZETIMIBE

 

Ezetimibe was classified as Class C pregnancy category; Class C meant that there is a lack of adequate studies in pregnant women, but animal studies have demonstrated a risk to the fetus (78). Animal studies have found that ezetimibe crosses the placenta. At levels higher than those achieved with human doses, there appears to be a slightly increased risk of skeletal abnormalities in rats and rabbits. Therefore, this agent is not recommended for use during pregnancy. If used prior to pregnancy, ezetimibe should be discontinued prior to attempting to become pregnant (98).The 2011 NLA Familial Hypercholesterolemia guidelines state that ezetimibe should be stopped at least 4 weeks before discontinuing contraception for women with familial hypercholesterolemia who are planning on conceiving and should not be used during pregnancy and lactation (97). 

 

PCSK9 INHIBITORS (EVOLOCUMAB, ALIROCUMAB, AND INCLISIRAN)

 

Evolocumab and alirocumab, have not been tested for safety during pregnancy so their role in dyslipidemia treatment in pregnancy is unclear. While older medications were labeled with pregnancy categories, such as A, B, C, D, and X, the FDA has removed these labels for all prescription medications approved after 2015 so there is no pregnancy classification for PCSK-9 inhibitors. The FDA drug package insert for evolocumab and alirocumab describe that monoclonal antibodies are unlikely to cross the placenta in the first trimester, but may cross

the placenta near term, in the second and third trimester.

 

Inclisiran, a small interfering RNA that targets hepatic PCSK9 synthesis, has been shown to significantly lower LDL-C levels. Given its infrequent dosing regimen, it could hypothetically be used before conception and immediately afterward though further trials and outcome data are needed (99). The product labeling states that there is no available data on its use in pregnant patients, although animal reproduction studies have shown no adverse developmental effects.

 

PCSK9 inhibitors are not approved for use in pregnancy nor currently recommended.

 

BEMPEDOIC ACID

 

Bempedoic acid, an inhibitor of ATP citrate lyase (an enzyme in the cholesterol synthesis pathway), is a lipid-lowering therapy shown to reduce the levels of LDL-C. Per the product labeling by the FDA, there is no available data on its use in pregnant women though animal reproduction studies did not show teratogenicity in rat and rabbit models (package insert). It is not recommended that bempedoic acid be taken during breastfeeding. They suggest discontinuing bempedoic acid when pregnancy is recognized, unless the benefits of therapy outweigh the potential risks to the fetus. Based on the mechanism of action of bempedoic acid may cause fetal harm.

 

EVINACUMAB

 

No data are available on use during pregnancy (96). Based on animal studies, exposure during pregnancy may lead to fetal harm. Evinacumab is a monoclonal antibody and human immune globulin are known to cross the placental barrier (96). Therefore, evinacumab could be transmitted from the mother to the developing fetus.

 

LOMITAPIDE

 

This drug is contraindicated during pregnancy due to the risk of fetal toxicity (formerly Class X pregnancy category) (100).

 

FIBRATES

 

While fibrates were classified as the Class C pregnancy category, they can be considered later on in pregnancy depending on the risk vs. benefit discussion. The AHA Scientific Statement for Cardiovascular Considerations in Caring for Pregnant Patients proposes the consideration of fenofibrate or gemfibrozil in the second trimester if triglycerides are >500 mg/dL despite lifestyle modifications (101). The AHA/ American College of Obstetricians and Gynecologists (ACOG) Presidential Advisory states that pregnant patients with a history of pancreatitis may benefit from the use of fenofibrate when triglyceride levels are >1000 mg/dL (66). The use of fibrates during the second trimester is after embryogenesis occurs reducing the risk. Studies in animals have found no increased risk of congenital malformations (98).

 

NIACIN

 

Niacin was classified in the Class C pregnancy category. Niacin should not be used during pregnancy and lactation.

 

OMEGA-3-FATTY ACIDS

 

Omega-3 fatty acids are widely used albeit without controlled clinical trials during pregnancy.

Studies are limited on the use of omega-3 fatty acids for dyslipidemia management during pregnancy. In one study of 341 pregnant women, omega-3 fatty acids in the form of 10 mL cod liver oil given daily until 3 months after delivery increased docosahexaenoic acid (DHA) levels in both maternal and infant plasma while also reducing maternal plasma lipid levels. Most other medications used to treat hypertriglyceridemia are not considered safe during pregnancy, but omega-3 fatty acids are considered safe as most prenatal vitamins and baby formula

contain DHA. Omega-3 fatty acids can potentially be utilized for their triglyceride-lowering effect, but evidence is only based on a small number of case reports (102). Prescription omega-3-fatty acids are not approved for use during pregnancy.

 

VOLANESORSEN

 

No data are available on use of volanesorsen during pregnancy. If used prior to pregnancy, volanesoren should be discontinued one month before attempting conception (103). This drug is approved in Europe but not in the U.S.

 

SUMMARY

 

Several recent reviews have provided information on the use of lipid lowering drugs during pregnancy and breast feeding (96,104,105). The class of evidence and levels of evidence for lipid lowering drugs are not strong for any of the options given a lack of definitive studies regarding efficacy and safety.  

 

Plasmapheresis and Lipoprotein Apheresis

 

In patients with severe elevations in triglyceride levels with pancreatitis or who are at high risk for pancreatitis plasmapheresis has been employed to rapidly and safely decrease triglyceride levels (81,106). Plasmapheresis should be considered early in asymptomatic pregnant women with fasting triglyceride levels >1000 mg/dL or in pregnant women with clinical signs and symptoms of pancreatitis and triglyceride levels >500 mg/dL despite maximal lifestyle changes and pharmacologic therapy.

 

Lipoprotein apheresis can be safely used during pregnancy and may be beneficial for some women with severely elevated LDL-C levels (85,106,107). An expert American College of Cardiology expert committee suggests consideration of lipoprotein apheresis in pregnant patients with homozygous familial hypercholesterolemia and patients with severe heterozygous familial hypercholesterolemia and an LDL-C ≥ 300 mg/dL despite lifestyle therapy (96). In patients with familial hypercholesterolemia, ASCVD, and pregnancy, lipoprotein apheresis may be considered when the LDL-C ≥190 mg/dL.

 

MANAGEMENT OF PREGNANT WOMEN WITH PRE-EXISTING LIPID ABNORMALITIES

  

Patients with the following disorders are best managed using a team approach that includes a lipid and maternal-fetal specialist. Persons with these disorders should be provided with genetic counseling in addition to multi-specialty team management and pregnancy offers an important opportunity to address the impact in families and how to recognize, prevent, and treat these disorders.

 

Elevated LDL-C Levels Including Homozygous and Heterozygous Familial Hypercholesterolemia

 

In women with familial hypercholesterolemia the percent increase in LDL-C levels during pregnancy is similar to that observed in women with a normal lipid profile prior to pregnancy even though the baseline LDL-C levels are much higher in women with familial hypercholesterolemia (92). Despite the markedly higher LDL-C levels in women with familial hypercholesterolemia the prevalence of hypertension, duration of gestation, and fetal body weight were similar between patients with familial hypercholesterolemia and women without dyslipidemia before pregnancy (92).

 

Decisions to use lipid-lowering medications involve a risk-benefit decision. In patients at very high risk of a heart attack or stroke, such as individuals with homozygous familial hypercholesterolemia and those who have clinical ASCVD the benefits of therapy may outweigh the risks of therapy. If the patient and physician elect to continue or add LDL-C lowering medications it is recommended that these be used after the first trimester of pregnancy if possible. Additionally, the hydrophilic statin, pravastatin, would be a good choice if one elects to use a statin (108). If the patient is on lipoprotein apheresis this can be continued during pregnancy or initiated if available.

 

Familial Chylomicronemia Syndrome (TG> 500mg/dL)

 

This is managed primarily with a very low-fat diet (<20 g total fat/d or <15% total calories) that requires consultation with an expert in nutritional advice to ensure adequate caloric intake during pregnancy and sufficient vitamins. Medium-chain triglycerides can help provide calories and make the very low-fat diet tolerable. In the third trimester as triglyceride levels increase hospitalization with parenteral feeding has been employed. In patients with familial chylomicronemia syndrome drug therapy often is not beneficial but one can consider omega-3 fatty acids in high doses. In patients with episodes of pancreatitis or with very high triglyceride levels at high risk for pancreatitis plasmapheresis can be beneficial.

 

Multifactorial Chylomicronemia Syndrome (TG> 500mg/dL)

 

This disorder is typically due to a genetic predisposition to high triglyceride levels combined with secondary factors that increase triglyceride levels into the range that causes pancreatitis. Therefore, one should try to control secondary disorders (Table 3) and if possible, stop drugs that increase triglyceride levels (Table 4). Additionally, a low-fat diet, avoidance of simple sugars and alcohol, and exercise can be helpful.

 

Table 3. Disorders Associated with an Increase in Triglyceride Levels

Obesity

Alcohol intake

High simple carbohydrate diet

Diabetes

Metabolic syndrome

Polycystic ovary syndrome

Hypothyroidism

Chronic renal failure

Nephrotic syndrome

Inflammatory diseases (Rheumatoid arthritis, Lupus, psoriasis, etc.)

Infections

Acute stress (myocardial infarctions, burns, etc.)

HIV

Cushing’s syndrome

Growth hormone deficiency

Lipodystrophy

Glycogen Storage disease

Acute hepatitis

Monoclonal gammopathy

 

Table 4. Drugs That Increase Triglyceride Levels

Alcohol

Oral Estrogens

Tamoxifen/Raloxifene

Glucocorticoids

Retinoids

Beta-blockers

Thiazide diuretics

Loop diuretics

Protease Inhibitors

Cyclosporine, sirolimus, and tacrolimus

Atypical antipsychotics

Bile acid sequestrants

L-asparaginase

Androgen deprivation therapy

Cyclophosphamide

Alpha-interferon

Propofol

 

If these are not successful in keeping triglyceride levels in a safe range treatment with omega-3- acids and the addition of fenofibrate later in pregnancy is indicated. If despite therapy there is an episode of pancreatitis due to poorly controlled triglyceride levels or very high triglyceride levels at high risk for pancreatitis plasmapheresis can be employed.

 

Patients with Moderate Hypertriglyceridemia (TG 200-500mg/dL)

 

These patients should be treated the same as patients with the multifactorial chylomicronemia syndrome described above. Diet therapy, treatment of secondary disorders, and stopping, if possible, drugs that increase triglyceride levels can frequently prevent an increase in triglycerides into the range that causes pancreatitis. If this approach is not successful treatment with omega-3- acids and the addition of fenofibrate later in pregnancy is indicated if the triglyceride levels are greater than 500mg/dL.

 

Patients with Very High Lp(a) Levels

 

High Lp(a) may promote atherosclerosis as well as thrombosis by affecting fibrinolysis, inflammation, endothelial function, macrophage lipid uptake, and oxidative stress. Lipoprotein(a) level correlates with the severity of preeclampsia and Lp(a) may be involved in the pathogenesis of preeclampsia (109). Further studies are required to determine if Lp(a) has a detrimental role during pregnancy and whether therapies that lower Lp(a) levels are beneficial.   To prevent pre-eclampsia aspirin therapy is recommended and can be considered for persons with high Lp(a).

 

Patients with Monogenic Disorders Causing Hypobetalipoproteinemia (i.e., Low LDL-C Levels)

 

There are a number of causes of low LDL-C including secondary factors such as a strict vegan diet, malnutrition, malabsorption, hyperthyroidism, malignancy, and chronic liver disease, polygenic disorders due to small effect variants in a number of genes, and several monogenic disorders (110). Amongst the monogenic disorders only the most severe are associated with pregnancy issues including bi-allelic FHBL-SD1 due to mutations in microsomal triglyceride transfer protein (abetalipoproteinemia), bi-allelic FHBL-SD2 due to mutations in Apo B, and bi-allelic FHBL-SD3 due to mutations in SAR1B (chylomicron retention disorder) (111). These disorders are characterized by very low LDL-C levels and malabsorption (111). The dietary treatment of these individuals is summarized in table 5 (111). Other monogenic disorders causing low LDL-C levels, such as mono-allelic FHBL-SD2 due to mutations in Apo B (familial hypobetalipoproteinemia), bi-allelic FHBL-EC1 due to mutations in ANGPTL3 (combined hypolipidemia), and bi-allelic FHBL-EC2 due to mutations in PCSK9 do not cause major issues during pregnancy because they do not lead to malabsorption.

 

Table 5. Treatment of Patients with Severe Monogenic Hypobetalipoproteinemia (Low LDL-C)

Low-fat diet- Less than 10-15% (<15 g/day) of total daily calories. Can be adjusted depending upon the tolerance

Essential fatty acids- 2-4% daily caloric intake (alpha-linolenic acid/linoleic acid)

DHA and EPA- Supplementation may be considered depending on the diet

Vitamin E- 100-300 IU/kg/day

Vitamin A- 100-400 IU/kg/day

Vitamin D- 800-1200 IU/day

Vitamin K- 5-35 mg/week

Dosing of vitamins A, D, and K can be tailored to plasma vitamin A/β-carotene levels, 25-hydroxy vitamin D levels, and INR reference intervals.

 

Preconception and pregnancy medical counseling, including genetic counseling, should be provided. Because patients with these disorders are very uncommon ideally these patients should be cared for in specialty clinics. During pregnancy, one must balance the need to limit fat intake and the need to increase caloric intake. Consultation with a dietician is important. Supplementation with medium-chain triglycerides can be used to increase caloric intake if needed (111). Because maternal serum DHA concentrations have been linked to neurocognitive and anti-inflammatory benefits supplementation with DHA (1-3 g/day) can be utilized to increase plasma concentrations (111). Additionally, to prevent neural tube defects in pregnancy, a daily supplement of 400-800 µg folic acid is also recommended. The dosing of vitamins A, D, and K should be adjusted as indicated by plasma vitamin A/β-carotene levels, 25-hydroxy vitamin D levels, and INR reference intervals (111). In normal individuals, excess vitamin A can cause toxicity during pregnancy and therefore it is recommended to set the vitamin A goal at the lower limit of normal levels, which may decrease the dose by 50% during pregnancy (111). Vitamin E supplements should be continued during pregnancy as vitamin E deficiency has been shown to increase miscarriages (111)   

 

Progesterone production during pregnancy may be reduced during pregnancy and it is recommended to monitor progesterone levels throughout pregnancy and consider the use of exogenous progesterone if levels are low (111).

 

CONSIDERATIONS FOR BREASTFEEDING

 

The postpartum period is a critical time requiring proactive counseling and shared decision-making regarding plans for lactation. Enabling women to breastfeed is a public health priority because, on a population level, interruption of lactation is associated with adverse health outcomes for the woman and her child, including higher maternal risks of breast cancer, ovarian cancer, diabetes, hypertension, and heart disease, and greater infant risks of infectious disease, sudden infant death syndrome, and metabolic disease. Contraindications to breastfeeding are few. Most medications and vaccinations are safe for use during breastfeeding, with few exceptions. Breastfeeding confers medical, economic, societal, and environmental advantages; however, each woman is uniquely qualified to make an informed decision surrounding infant feeding.

 

Risks vs. benefits need to be considered to determine the optimal management of lipid disorders, as all lipid-lowering medications are contraindicated during pregnancy and remain contraindicated during breastfeeding. Breastfeeding has enormous benefits and is encouraged. Medications taken by the mother can transfer into breast milk through passive diffusion or active transport by membrane proteins and therefore expose the baby to the medication. Therefore, continuing breastfeeding given its benefits in metabolic health needs to be balanced with stopping breastfeeding to resume lipid-lowering therapies and the progression of atherosclerosis (112). Shared decision-making and discussions of risks and benefits of both time without statin treatment and breastfeeding vs. treatment during pregnancy and lactation are best initiated before delivery and every visit in the fourth-trimester period (after delivery) (113,114). The LactMed database (National Institute of Child Health and Development) contains information on drugs and other chemicals to which breastfeeding mothers may be exposed. It includes information on the levels of such substances in breast milk and infant blood and the possible adverse effects in the nursing infant. Suggested therapeutic alternatives to those drugs are provided, where appropriate. All data are derived from the scientific literature and fully referenced. A peer review panel reviews the data to assure scientific validity and currency. The consensus opinion is that women taking a statin should not breastfeed because of a concern with disruption of infant lipid metabolism. However, others have argued that children homozygous for familial hypercholesterolemia are treated with statins beginning at 1 year of age, that statins have low oral bioavailability, and that risks to the breastfed infant are low, especially with pravastatin and rosuvastatin.

 

In a recent systematic review of 33 articles from 15 randomized controlled trials limited evidence suggests that omega-3 fatty acid supplementation during pregnancy may result in favorable cognitive development in the child. There was insufficient evidence to evaluate the effects of omega-3 fatty acid supplementation during pregnancy and/or lactation on other developmental outcomes (115).

 

CONCLUSION

 

There is an increase in lipid levels in normal gestation. Dyslipidemia in pregnancy beyond physiologic levels is associated with adverse pregnancy outcomes. Adverse pregnancy events enhance the risk of clinical ASCVD events in later life. Screening for and adequately addressing atherogenic dyslipidemia before and during pregnancy is a priority. Major barriers to the management of hyperlipidemia during pregnancy and the postpartum period include limited studies in pregnant patients. Many therapeutic agents are categorized as contraindicated without adequate evidence. Future research is needed to allow for evidence-based decisions to guide therapeutic options. Pregnancy is a unique opportunity for multidisciplinary and collaborative care across various specialties to improve rates of screening and optimize the long-term cardiovascular health of women. Many women are overwhelmed post-partum. They need to be encouraged to prioritize ASCVD risk reduction as integral to their care. Improving access to quality preventive care is still in need of improvement.

 

REFERENCES

 

  1. Woollett LA. Where Does Fetal and Embryonic Cholesterol Originate and What Does It Do? Annual Review of Nutrition 2008; 28:97-114
  2. Herman GE. Disorders of cholesterol biosynthesis: prototypic metabolic malformation syndromes. Human Molecular Genetics 2003; 12:75R-88
  3. Kelley RI. Inborn errors of cholesterol biosynthesis. Adv Pediatr 2000; 47:1-53
  4. Woollett LA. Maternal cholesterol in fetal development: transport of cholesterol from the maternal to the fetal circulation. The American Journal of Clinical Nutrition 2005; 82:1155-1161
  5. Yoshida S, Wada Y. Transfer of maternal cholesterol to embryo and fetus in pregnant mice. Journal of Lipid Research 2005; 46:2168-2174
  6. Vuorio AF, Miettinen TA, Turtola H, Oksanen H, Gylling H. Cholesterol metabolism in normal and heterozygous familial hypercholesterolemic newborns. Journal of Laboratory and Clinical Medicine 2002; 140:35-42
  7. Linck LM, Hayflick SJ, Lin DS, Battaile KP, Ginat S, Burlingame T, Gibson KM, Honda M, Honda A, Salen G, Tint GS, Connor WE, Steiner RD. Fetal demise with Smith-Lemli-Opitz syndrome confirmed by tissue sterol analysis and the absence of measurable 7-dehydrocholesterol ?7-reductase activity in chorionic villi. Prenatal Diagnosis 2000; 20:238-240
  8. Nowaczyk MgJM, Farrell SA, Sirkin WL, Velsher L, Krakowiak PA, Waye JS, Porter FD. Smith-Lemli-Opitz (RHS) syndrome: holoprosencephaly and homozygous IVS8-1G?C genotype. American Journal of Medical Genetics 2001; 103:75-80
  9. Spellacy WN, Ashbacher LV, Harris GK, Buhi WC. Total cholesterol content in maternal and umbilical vessels in term pregnancies. Obstet Gynecol 1974; 44:661-665
  10. Woollett LA, Heubi JE. Fetal and Neonatal Cholesterol Metabolism. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Hofland J, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2020.
  11. Woollett LA. Fetal lipid metabolism. Frontiers in Bioscience 2001; 6:d536-545
  12. Nagasaka H, Chiba H, Kikuta H, Akita H, Takahashi Y, Yanai H, Hui SP, Fuda H, Fujiwara H, Kobayashi K. Unique character and metabolism of high density lipoprotein (HDL) in fetus. Atherosclerosis 2002; 161:215-223
  13. Sreckovic I, Birner-Gruenberger R, Obrist B, Stojakovic T, Scharnagl H, Holzer M, Scholler M, Philipose S, Marsche G, Lang U, Desoye G, Wadsack C. Distinct composition of human fetal HDL attenuates its anti-oxidative capacity. Biochim Biophys Acta 2013; 1831:737-746
  14. Schmid KE, Davidson WS, Myatt L, Woollett LA. Transport of cholesterol across a BeWo cell monolayer: implications for net transport of sterol from maternal to fetal circulation. J Lipid Res 2003; 44:1909-1918
  15. Strahlhofer-Augsten M, Schliefsteiner C, Cvitic S, George M, Lang-Olip I, Hirschmugl B, Marsche G, Lang U, Novakovic B, Saffery R, Desoye G, Wadsack C. The Distinct Role of the HDL Receptor SR-BI in Cholesterol Homeostasis of Human Placental Arterial and Venous Endothelial Cells. Int J Mol Sci 2022; 23
  16. Bresnitz W, Lorca RA. Potassium Channels in the Uterine Vasculature: Role in Healthy and Complicated Pregnancies. Int J Mol Sci 2022; 23
  17. Burton GJ, Fowden AL, Thornburg KL. Placental Origins of Chronic Disease. Physiol Rev 2016; 96:1509-1565
  18. Brett KE, Ferraro ZM, Yockell-Lelievre J, Gruslin A, Adamo KB. Maternal-fetal nutrient transport in pregnancy pathologies: the role of the placenta. Int J Mol Sci 2014; 15:16153-16185
  19. Harris WS, Baack ML. Beyond building better brains: bridging the docosahexaenoic acid (DHA) gap of prematurity. J Perinatol 2015; 35:1-7
  20. Wang Y, Chen Z, Zhang F. Association between maternal lipid levels during pregnancy and delivery of small for gestational age: A systematic review and meta-analysis. Front Pediatr 2022; 10:934505
  21. Higa R, Jawerbaum A. Intrauterine effects of impaired lipid homeostasis in pregnancy diseases. Curr Med Chem 2013; 20:2338-2350
  22. Woo JG, Melchior JT, Swertfeger DK, Remaley AT, Sise EA, Sosseh F, Welge JA, Prentice AM, Davidson WS, Moore SE, Woollett LA. Lipoprotein subfraction patterns throughout gestation in The Gambia: changes in subfraction composition and their relationships with infant birth weights. Lipids Health Dis 2023; 22:19
  23. Wild RA, Weedin E, Cox K, Zhao YD, Wrenn DS, Lopez D, Wooten CJ, Melendez QM, Myers D, Hansen KR. Proprotein Convertase Subtilisin Kexin 9 (PCSK9) and nonHDL particles rise during normal pregnancy and differ by BMI. J Clin Lipidol 2022; 16:483-490
  24. Wiznitzer A, Mayer A, Novack V, Sheiner E, Gilutz H, Malhotra A, Novack L. Association of lipid levels during gestation with preeclampsia and gestational diabetes mellitus: a population-based study. American journal of obstetrics and gynecology 2009; 201:482.e481-482.e4828
  25. Piechota W, Staszewski A. Reference ranges of lipids and apolipoproteins in pregnancy. Eur J Obstet Gynecol Reprod Biol 1992; 45:27-35
  26. S B. Maternal, Fetal, & Neonatal Physiology. Elsevier.
  27. Herrera E. Lipid Metabolism in Pregnancy and its Consequences in the Fetus and Newborn. Endocrine 2002; 19:43-56
  28. Loke DFM, Viegas OAC, Kek LP, Rauff M, Thai AC, Ratnam SS. Lipid Profiles during and after Normal Pregnancy. Gynecologic and Obstetric Investigation 1991; 32:144-147
  29. Alvarez JJ, Montelongo A, Iglesias A, Lasunción MA, Herrera E. Longitudinal study on lipoprotein profile, high density lipoprotein subclass, and postheparin lipases during gestation in women. Journal of Lipid Research1996; 37:299-308
  30. Woollett LA, Catov JM, Jones HN. Roles of maternal HDL during pregnancy. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159106
  31. Sattar N, Clark P, Greer IA, Shepherd J, Packard CJ. Lipoprotein (a) levels in normal pregnancy and in pregnancy complicated with pre-eclampsia. Atherosclerosis 2000; 148:407-411
  32. Brizzi P, Tonolo G, Esposito F, Puddu L, Dessole S, Maioli M, Milia S. Lipoprotein metabolism during normal pregnancy. Am J Obstet Gynecol 1999; 181:430-434
  33. Basaran A. Pregnancy-induced hyperlipoproteinemia: review of the literature. Reprod Sci 2009; 16:431-437
  34. Rymer J, Constable S, Lumb P, Crook M. Serum lipoprotein (A) and apolipoproteins during pregnancy and postpartum in normal women. J Obstet Gynaecol 2002; 22:256-259
  35. Mazurkiewicz JC, Watts GF, Warburton FG, Slavin BM, Lowy C, Koukkou E. Serum lipids, lipoproteins and apolipoproteins in pregnant non-diabetic patients. J Clin Pathol 1994; 47:728-731
  36. Manten GT, Franx A, van der Hoek YY, Hameeteman TM, Voorbij HA, Smolders HC, Westers P, Visser GH. Changes of plasma lipoprotein(a) during and after normal pregnancy in Caucasians. J Matern Fetal Neonatal Med 2003; 14:91-95
  37. Zechner R, Desoye G, Schweditsch MO, Pfeiffer KP, Kostner GM. Fluctuations of plasma lipoprotein-A concentrations during pregnancy and post partum. Metabolism 1986; 35:333-336
  38. Napso T, Yong HEJ, Lopez-Tello J, Sferruzzi-Perri AN. The Role of Placental Hormones in Mediating Maternal Adaptations to Support Pregnancy and Lactation. Front Physiol 2018; 9:1091
  39. Brown MS, Goldstein JL. Selective versus total insulin resistance: a pathogenic paradox. Cell Metab 2008; 7:95-96
  40. Applebaum DM, Goldberg AP, Pykälistö OJ, Brunzell JD, Hazzard WR. Effect of estrogen on post-heparin lipolytic activity. Selective decline in hepatic triglyceride lipase. J Clin Invest 1977; 59:601-608
  41. Feingold KR, Wiley T, Moser AH, Lear SR, Wiley MH. De novo cholesterogenesis in pregnancy. J Lab Clin Med 1983; 101:256-263
  42. Reichen J, Karlaganis G, Kern F. Cholesterol synthesis in the perfused liver of pregnant hamsters. Journal of Lipid Research 1987; 28:1046-1052
  43. Habibi N, Mousa A, Tay CT, Khomami MB, Patten RK, Andraweera PH, Wassie M, Vandersluys J, Aflatounian A, Bianco-Miotto T, Zhou SJ, Grieger JA. Maternal metabolic factors and the association with gestational diabetes: A systematic review and meta-analysis. Diabetes Metab Res Rev 2022; 38:e3532
  44. Ryckman KK, Spracklen CN, Smith CJ, Robinson JG, Saftlas AF. Maternal lipid levels during pregnancy and gestational diabetes: a systematic review and meta-analysis. BJOG 2015; 122:643-651
  45. Pathirana MM, Lassi Z, Ali A, Arstall M, Roberts CT, Andraweera PH. Cardiovascular risk factors in women with previous gestational diabetes mellitus: A systematic review and meta-analysis. Rev Endocr Metab Disord2021; 22:729-761
  46. Kramer CK, Campbell S, Retnakaran R. Gestational diabetes and the risk of cardiovascular disease in women: a systematic review and meta-analysis. Diabetologia 2019; 62:905-914
  47. Arya S, Hansen KR, Peck JD, Wild RA. Metabolic syndrome in obesity: treatment success and adverse pregnancy outcomes with ovulation induction in polycystic ovary syndrome. Am J Obstet Gynecol 2021; 225:280.e281-280.e211
  48. Poornima IG, Indaram M, Ross JD, Agarwala A, Wild RA. Hyperlipidemia and risk for preclampsia. J Clin Lipidol 2022; 16:253-260
  49. Wang J, Moore D, Subramanian A, Cheng KK, Toulis KA, Qiu X, Saravanan P, Price MJ, Nirantharakumar K. Gestational dyslipidaemia and adverse birthweight outcomes: a systematic review and meta-analysis. Obes Rev 2018; 19:1256-1268
  50. Mahindra MP, Sampurna MTA, Mapindra MP, Sutowo Putri AM. Maternal lipid levels in pregnant women without complications in developing risk of large for gestational age newborns: a study of meta-analysis. F1000Res 2020; 9:1213
  51. Vrijkotte TGM, Krukziener N, Hutten BA, Vollebregt KC, van Eijsden M, Twickler MB. Maternal Lipid Profile During Early Pregnancy and Pregnancy Complications and Outcomes: The ABCD Study. The Journal of Clinical Endocrinology &amp; Metabolism 2012; 97:3917-3925
  52. Knopp RH, Magee MS, Walden CE, Bonet B, Benedetti TJ. Prediction of Infant Birth Weight by GDM Screening Tests: Importance of plasma triglyceride. Diabetes Care 1992; 15:1605-1613
  53. Nolan CJ, Riley SF, Sheedy MT, Walstab JE, Beischer NA. Maternal Serum Triglyceride, Glucose Tolerance, and Neonatal Birth Weight Ratio in Pregnancy: A study within a racially heterogeneous population. Diabetes Care 1995; 18:1550-1556
  54. Schaefer-Graf UM, Graf K, Kulbacka I, Kjos SL, Dudenhausen J, Vetter K, Herrera E. Maternal lipids as strong determinants of fetal environment and growth in pregnancies with gestational diabetes mellitus. Diabetes care2008; 31:1858-1863
  55. Di Cianni G, Miccoli R, Volpe L, Lencioni C, Ghio A, Giovannitti MG, Cuccuru I, Pellegrini G, Chatzianagnostou K, Boldrini A, Del Prato S. Maternal triglyceride levels and newborn weight in pregnant women with normal glucose tolerance. Diabet Med 2005; 22:21-25
  56. Wang X, Guan Q, Zhao J, Yang F, Yuan Z, Yin Y, Fang R, Liu L, Zuo C, Gao L. Association of maternal serum lipids at late gestation with the risk of neonatal macrosomia in women without diabetes mellitus. Lipids Health Dis 2018; 17:78
  57. Ye K, Bo QL, Du QJ, Zhang D, Shen Y, Han YP, Li YB, Li Y, Hu CL, Li L. Maternal serum lipid levels during late pregnancy and neonatal body size. Asia Pac J Clin Nutr 2015; 24:138-143
  58. Spracklen CN, Smith CJ, Saftlas AF, Robinson JG, Ryckman KK. Maternal hyperlipidemia and the risk of preeclampsia: a meta-analysis. Am J Epidemiol 2014; 180:346-358
  59. Gallos ID, Sivakumar K, Kilby MD, Coomarasamy A, Thangaratinam S, Vatish M. Pre-eclampsia is associated with, and preceded by, hypertriglyceridaemia: a meta-analysis. BJOG: An International Journal of Obstetrics &amp; Gynaecology 2013; 120:1321-1332
  60. Tesfa E, Nibret E, Munshea A. Maternal lipid profile and risk of pre-eclampsia in African pregnant women: A systematic review and meta-analysis. PLoS One 2020; 15:e0243538
  61. Enquobahrie D. Maternal plasma lipid concentrations in early pregnancy and risk of preeclampsia*1. American Journal of Hypertension 2004; 17:574-581
  62. Vaught AJ, Boyer T, Ziogos E, Amat-Codina N, Minhas A, Darwin K, Debrosse A, Fedarko N, Burd I, Baschat A, Sharma G, Hays AG, Zakaria S, Leucker TM. The role of proprotein convertase subtillisin/kexin type 9 in placental salvage and lipid metabolism in women with preeclampsia. Placenta 2023; 132:1-6
  63. Wild RA, Edwards RK, Zhao D, Hansen KR, Kim AS, Wrenn DS. Highly Atherogenic Lipid Particles are Associated with Preeclampsia After Successful Fertility Treatment for Obese Women who have Unexplained Infertility. Reprod Sci 2023;
  64. Alonso-Ventura V, Li Y, Pasupuleti V, Roman YM, Hernandez AV, Perez-Lopez FR. Effects of preeclampsia and eclampsia on maternal metabolic and biochemical outcomes in later life: a systematic review and meta-analysis. Metabolism 2020; 102:154012
  65. McDonald SD, Malinowski A, Zhou Q, Yusuf S, Devereaux PJ. Cardiovascular sequelae of preeclampsia/eclampsia: a systematic review and meta-analyses. Am Heart J 2008; 156:918-930
  66. Brown MC, Best KE, Pearce MS, Waugh J, Robson SC, Bell R. Cardiovascular disease risk in women with pre-eclampsia: systematic review and meta-analysis. Eur J Epidemiol 2013; 28:1-19
  67. Jiang S, Jiang J, Xu H, Wang S, Liu Z, Li M, Liu H, Zheng S, Wang L, Fei Y, Li X, Ding Y, Wang Z, Yu Y. Maternal dyslipidemia during pregnancy may increase the risk of preterm birth: A meta-analysis. Taiwan J Obstet Gynecol 2017; 56:9-15
  68. Napoli C, Ackah E, De Nigris F, Del Soldato P, D'Armiento FP, Crimi E, Condorelli M, Sessa WC. Chronic treatment with nitric oxide-releasing aspirin reduces plasma low-density lipoprotein oxidation and oxidative stress, arterial oxidation-specific epitopes, and atherogenesis in hypercholesterolemic mice. Proc Natl Acad Sci U S A 2002; 99:12467-12470
  69. Palinski W, D’Armiento FP, Witztum JL, de Nigris F, Casanada F, Condorelli M, Silvestre M, Napoli C. Maternal Hypercholesterolemia and Treatment During Pregnancy Influence the Long-Term Progression of Atherosclerosis in Offspring of Rabbits. Circulation Research 2001; 89:991-996
  70. Palinski W, Napoli C. The fetal origins of atherosclerosis: maternal hypercholesterolemia, and cholesterol-lowering or antioxidant treatment during pregnancy influence in utero programming and postnatal susceptibility to atherogenesis. FASEB J 2002; 16:1348-1360
  71. Schiano C, D'Armiento M, Franzese M, Castaldo R, Saccone G, de Nigris F, Grimaldi V, Soricelli A, D'Armiento FP, Zullo F, Napoli C. DNA Methylation Profile of the SREBF2 Gene in Human Fetal Aortas. J Vasc Res 2022; 59:61-68
  72. Cacciatore F, Bruzzese G, Abete P, Russo G, Palinski W, Napoli C. Maternal hypercholesterolaemia during pregnancy affects severity of myocardial infarction in young adults. Eur J Prev Cardiol 2022; 29:758-765
  73. Stefulj J, Panzenboeck U, Becker T, Hirschmugl B, Schweinzer C, Lang I, Marsche G, Sadjak A, Lang U, Desoye G, Wadsack C. Human endothelial cells of the placental barrier efficiently deliver cholesterol to the fetal circulation via ABCA1 and ABCG1. Circ Res 2009; 104:600-608
  74. Mendelson MM, Lyass A, O'Donnell CJ, D'Agostino RB, Sr., Levy D. Association of Maternal Prepregnancy Dyslipidemia With Adult Offspring Dyslipidemia in Excess of Anthropometric, Lifestyle, and Genetic Factors in the Framingham Heart Study. JAMA Cardiol 2016; 1:26-35
  75. Øyri LKL, Bogsrud MP, Christensen JJ, Ulven SM, Brantsæter AL, Retterstøl K, Brekke HK, Michelsen TM, Henriksen T, Roeters van Lennep JE, Magnus P, Veierød MB, Holven KB. Novel associations between parental and newborn cord blood metabolic profiles in the Norwegian Mother, Father and Child Cohort Study. BMC Med 2021; 19:91
  76. Ma J, Rebholz CM, Braun KVE, Reynolds LM, Aslibekyan S, Xia R, Biligowda NG, Huan T, Liu C, Mendelson MM, Joehanes R, Hu EA, Vitolins MZ, Wood AC, Lohman K, Ochoa-Rosales C, van Meurs J, Uitterlinden A, Liu Y, Elhadad MA, Heier M, Waldenberger M, Peters A, Colicino E, Whitsel EA, Baldassari A, Gharib SA, Sotoodehnia N, Brody JA, Sitlani CM, Tanaka T, Hill WD, Corley J, Deary IJ, Zhang Y, Schottker B, Brenner H, Walker ME, Ye S, Nguyen S, Pankow J, Demerath EW, Zheng Y, Hou L, Liang L, Lichtenstein AH, Hu FB, Fornage M, Voortman T, Levy D. Whole Blood DNA Methylation Signatures of Diet Are Associated With Cardiovascular Disease Risk Factors and All-Cause Mortality. Circ Genom Precis Med 2020; 13:e002766
  77. Venetkoski M, Joensuu J, Gissler M, Ylikorkala O, Mikkola TS, Savolainen-Peltonen H. Pre-eclampsia and cardiovascular risk: a long-term nationwide cohort study on over 120 000 Finnish women. BMJ Open 2022; 12:e064736
  78. Jacobson TA, Maki KC, Orringer CE, Jones PH, Kris-Etherton P, Sikand G, La Forge R, Daniels SR, Wilson DP, Morris PB, Wild RA, Grundy SM, Daviglus M, Ferdinand KC, Vijayaraghavan K, Deedwania PC, Aberg JA, Liao KP, McKenney JM, Ross JL, Braun LT, Ito MK, Bays HE, Brown WV, Underberg JA. National Lipid Association Recommendations for Patient-Centered Management of Dyslipidemia: Part 2. J Clin Lipidol 2015; 9:S1-122.e121
  79. Kenik J, Jean-Jacques M, Feinglass J. Explaining racial and ethnic disparities in cholesterol screening. Prev Med 2014; 65:65-69
  80. Satish P, Sadaf MI, Valero-Elizondo J, Grandhi GR, Yahya T, Zawahir H, Javed Z, Mszar R, Hanif B, Kalra A, Virani S, Cainzos-Achirica M, Nasir K. Heterogeneity in cardio-metabolic risk factors and atherosclerotic cardiovascular disease among Asian groups in the United States. Am J Prev Cardiol 2021; 7:100219
  81. Gupta M, Liti B, Barrett C, Thompson PD, Fernandez AB. Prevention and Management of Hypertriglyceridemia-Induced Acute Pancreatitis During Pregnancy: A Systematic Review. Am J Med 2022; 135:709-714
  82. Grundy SM, Stone NJ. 2018 American Heart Association/American College of Cardiology Multisociety Guideline on the Management of Blood Cholesterol: Primary Prevention. JAMA Cardiol 2019; 4:488-489
  83. Brown HL, Warner JJ, Gianos E, Gulati M, Hill AJ, Hollier LM, Rosen SE, Rosser ML, Wenger NK, American Heart A, the American College of O, Gynecologists. Promoting Risk Identification and Reduction of Cardiovascular Disease in Women Through Collaboration With Obstetricians and Gynecologists: A Presidential Advisory From the American Heart Association and the American College of Obstetricians and Gynecologists. Circulation 2018; 137:e843-e852
  84. Khoury J, Haugen G, Tonstad S, Frøslie KF, Henriksen T. Effect of a cholesterol-lowering diet during pregnancy on maternal and fetal Doppler velocimetry: the CARRDIP study. Am J Obstet Gynecol 2007; 196:549.e541-547
  85. Grundy SM, Stone NJ, Bailey AL, Beam C, Birtcher KK, Blumenthal RS, Braun LT, de Ferranti S, Faiella-Tommasino J, Forman DE, Goldberg R, Heidenreich PA, Hlatky MA, Jones DW, Lloyd-Jones D, Lopez-Pajares N, Ndumele CE, Orringer CE, Peralta CA, Saseen JJ, Smith SC, Jr., Sperling L, Virani SS, Yeboah J. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA Guideline on the Management of Blood Cholesterol: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 2019; 139:e1082-e1143
  86. Mach F, Baigent C, Catapano AL, Koskinas KC, Casula M, Badimon L, Chapman MJ, De Backer GG, Delgado V, Ference BA, Graham IM, Halliday A, Landmesser U, Mihaylova B, Pedersen TR, Riccardi G, Richter DJ, Sabatine MS, Taskinen MR, Tokgozoglu L, Wiklund O, Group ESCSD. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: lipid modification to reduce cardiovascular risk. Eur Heart J 2020; 41:111-188
  87. Karalis DG, Hill AN, Clifton S, Wild RA. The risks of statin use in pregnancy: A systematic review. Journal of Clinical Lipidology 2016; 10:1081-1090
  88. Edison RJ, Muenke M. Central nervous system and limb anomalies in case reports of first-trimester statin exposure. N Engl J Med 2004; 350:1579-1582
  89. Zarek J, Koren G. The fetal safety of statins: a systematic review and meta-analysis. J Obstet Gynaecol Can2014; 36:506-509
  90. Botha TC, Pilcher GJ, Wolmarans K, Blom DJ, Raal FJ. Statins and other lipid-lowering therapy and pregnancy outcomes in homozygous familial hypercholesterolaemia: A retrospective review of 39 pregnancies. Atherosclerosis 2018; 277:502-507
  91. Halpern DG, Weinberg CR, Pinnelas R, Mehta-Lee S, Economy KE, Valente AM. Use of Medication for Cardiovascular Disease During Pregnancy: JACC State-of-the-Art Review. J Am Coll Cardiol 2019; 73:457-476
  92. Amundsen AL, Khoury J, Iversen PO, Bergei C, Ose L, Tonstad S, Retterstol K. Marked changes in plasma lipids and lipoproteins during pregnancy in women with familial hypercholesterolemia. Atherosclerosis 2006; 189:451-457
  93. Toleikyte I, Retterstol K, Leren TP, Iversen PO. Pregnancy outcomes in familial hypercholesterolemia: a registry-based study. Circulation 2011; 124:1606-1614
  94. Dobert M, Varouxaki AN, Mu AC, Syngelaki A, Ciobanu A, Akolekar R, De Paco Matallana C, Cicero S, Greco E, Singh M, Janga D, Del Mar Gil M, Jani JC, Bartha JL, Maclagan K, Wright D, Nicolaides KH. Pravastatin Versus Placebo in Pregnancies at High Risk of Term Preeclampsia. Circulation 2021; 144:670-679
  95. Hirsch A, Rotem R, Ternovsky N, Hirsh Raccah B. Pravastatin and placental insufficiency associated disorders: A systematic review and meta-analysis. Front Pharmacol 2022; 13:1021548
  96. Writing Committee, Lloyd-Jones DM, Morris PB, Ballantyne CM, Birtcher KK, Covington AM, DePalma SM, Minissian MB, Orringer CE, Smith SC, Jr., Waring AA, Wilkins JT. 2022 ACC Expert Consensus Decision Pathway on the Role of Nonstatin Therapies for LDL-Cholesterol Lowering in the Management of Atherosclerotic Cardiovascular Disease Risk: A Report of the American College of Cardiology Solution Set Oversight Committee. J Am Coll Cardiol 2022; 80:1366-1418
  97. Ito MK, McGowan MP, Moriarty PM, National Lipid Association Expert Panel on Familial H. Management of familial hypercholesterolemias in adult patients: recommendations from the National Lipid Association Expert Panel on Familial Hypercholesterolemia. J Clin Lipidol 2011; 5:S38-45
  98. Liebeskind A, Thompson J, Wilson D. Reproductive Health and its Impact on Lipid Management in Adolescent and Young Adult Females. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Hofland J, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2022.
  99. Raal FJ, Kallend D, Ray KK, Turner T, Koenig W, Wright RS, Wijngaard PLJ, Curcio D, Jaros MJ, Leiter LA, Kastelein JJP, Investigators O-. Inclisiran for the Treatment of Heterozygous Familial Hypercholesterolemia. N Engl J Med 2020; 382:1520-1530
  100. Cefalu AB, D'Erasmo L, Iannuzzo G, Noto D, Giammanco A, Montali A, Zambon A, Forte F, Suppressa P, Giannini S, Barbagallo CM, Ganci A, Nardi E, Vernuccio F, Caldarella R, Ciaccio M, Arca M, Averna M. Efficacy and safety of lomitapide in familial chylomicronaemia syndrome. Atherosclerosis 2022; 359:13-19
  101. Mehta LS, Warnes CA, Bradley E, Burton T, Economy K, Mehran R, Safdar B, Sharma G, Wood M, Valente AM, Volgman AS, American Heart Association Council on Clinical C, Council on Arteriosclerosis T, Vascular B, Council on C, Stroke N, Stroke C. Cardiovascular Considerations in Caring for Pregnant Patients: A Scientific Statement From the American Heart Association. Circulation 2020; 141:e884-e903
  102. Helland IB, Saugstad OD, Saarem K, Van Houwelingen AC, Nylander G, Drevon CA. Supplementation of n-3 fatty acids during pregnancy and lactation reduces maternal plasma lipid levels and provides DHA to the infants. J Matern Fetal Neonatal Med 2006; 19:397-406
  103. Kolovou G, Kolovou V, Katsiki N. Volanesorsen: A New Era in the Treatment of Severe Hypertriglyceridemia. J Clin Med 2022; 11
  104. Kaur G, Gulati M. Considerations for treatment of lipid disorders during pregnancy and breastfeeding. Prog Cardiovasc Dis 2022; 75:33-39
  105. Grant JK, Snow S, Kelsey M, Rymer J, Schaffer AE, Patel MR, McGarrah RW, Pagidipati NJ, Shah NP. Lipid-Lowering Therapy in Women of Childbearing Age: a Review and Stepwise Clinical Approach. Curr Cardiol Rep2022; 24:1373-1385
  106. Russi G. Severe dyslipidemia in pregnancy: The role of therapeutic apheresis. Transfusion and Apheresis Science 2015; 53:283-287
  107. Blaha M, Veletova K, Blaha V, Lanska M, Zak P. Pregnancy in homozygous familial hypercholesterolemia-A case series. Ther Apher Dial 2022; 26 Suppl 1:89-96
  108. Graham DF, Raal FJ. Management of familial hypercholesterolemia in pregnancy. Curr Opin Lipidol 2021; 32:370-377
  109. Konrad E, Guralp O, Shaalan W, Elzarkaa AA, Moftah R, Alemam D, Malik E, Soliman AA. Correlation of elevated levels of lipoprotein(a), high-density lipoprotein and low-density lipoprotein with severity of preeclampsia: a prospective longitudinal study. J Obstet Gynaecol 2020; 40:53-58
  110. Shapiro MD, Feingold KR. Monogenic Disorders Causing Hypobetalipoproteinemia. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2022.
  111. Bredefeld C, Hussain MM, Averna M, Black DD, Brin MF, Burnett JR, Charriere S, Cuerq C, Davidson NO, Deckelbaum RJ, Goldberg IJ, Granot E, Hegele RA, Ishibashi S, Karmally W, Levy E, Moulin P, Okazaki H, Poinsot P, Rader DJ, Takahashi M, Tarugi P, Traber MG, Di Filippo M, Peretti N. Guidance for the diagnosis and treatment of hypolipidemia disorders. J Clin Lipidol 2022; 16:797-812
  112. Klevmoen M, Bogsrud MP, Retterstøl K, Svilaas T, Vesterbekkmo EK, Hovland A, Berge C, Roeters van Lennep J, Holven KB. Loss of statin treatment years during pregnancy and breastfeeding periods in women with familial hypercholesterolemia. Atherosclerosis 2021; 335:8-15
  113. ACOG Committee Opinion No. 736: Optimizing Postpartum Care. Obstet Gynecol 2018; 131:e140-e150
  114. ACOG Committee Opinion No. 756: Optimizing Support for Breastfeeding as Part of Obstetric Practice. Obstet Gynecol 2018; 132:e187-e196
  115. Nevins JEH, Donovan SM, Snetselaar L, Dewey KG, Novotny R, Stang J, Taveras EM, Kleinman RE, Bailey RL, Raghavan R, Scinto-Madonich SR, Venkatramanan S, Butera G, Terry N, Altman J, Adler M, Obbagy JE, Stoody EE, de Jesus J. Omega-3 Fatty Acid Dietary Supplements Consumed During Pregnancy and Lactation and Child Neurodevelopment: A Systematic Review. J Nutr 2021; 151:3483-3494

Genetic Disorders Causing Hypertriglyceridemia in Children and Adolescents

ABSTRACT

 

Primary disorders of lipid metabolism causing hypertriglyceridemia (HTG) result from genetic defects in triglyceride synthesis and metabolism. These disorders, with the exception of mutations in the lipoprotein lipase complex, are often unmasked by precipitating factors including obesity, diabetes, or medications. Physical findings can include eruptive, palmer, or tuberoeruptive xanthomas. Other lipid abnormalities may or may not be present. Each of the genetic causes of HTG is associated with an increased risk of developing recurrent pancreatitis; some may also increase the risk of premature cardiovascular disease. Appropriate management begins with proper recognition of the disorder. Pharmacotherapies for TG lowering, although not approved for use in children <18 years-of-age in the U.S.,are available and may be beneficial in select disorders. We review the genetic disorders causing HTG in children and adolescents, discuss their clinical presentation, associated complications, and management, and conclude with novel therapies in development.

 

INTRODUCTION

 

Triglycerides (TGs) constitute one of the major lipid groups. Excessive accumulation of TG in the blood leads to hypertriglyceridemia. TG concentrations of > 500 mg/dL account for <0.2% of the HTG cases in children, but when encountered should prompt consideration of mutations in the lipoprotein lipase (LPL) complex, termed the familial chylomicronemia syndrome (FCS) or the co-existence of genetic and secondary forms of HTG, termed the multifactorial chylomicronemia syndrome (MFCS), a far more common cause of severe HTG. Secondary causes of HTG include unrecognized or poorly controlled diabetes, obesity, metabolic syndrome, and medications (including atypical antipsychotics and estrogens) (Table 1). Appropriate management of the patient with HTG requires knowledge of the likely cause of the HTG, in order to prevent its complications. Our focus is to review the pathogenesis, genetics, presentation, and diagnosis of inherited HTG disorders in children and adolescents.

 

Table 1. Secondary Causes Hypertriglyceridemia in Children and Adolescents

Diet with excess calories, high glycemic load, and/or sucrose- or fructose-containing beverages

Endocrine Disorders (uncontrolled type 1 and type 2 diabetes mellitus, obesity, metabolic syndrome, hypothyroidism, hypercortisolism, lipodystrophies)

Medications (steroids, oral estrogen, second generation antipsychotics, antidepressants, retinoic acid derivatives, rosiglitazone, thiazide diuretics, beta-blockers, bile acid sequestrants, sirolimus, PEG-asparaginase, antiretroviral therapy)

Pregnancy

Renal disease (nephrotic syndrome, renal failure)

Liver disease (acute hepatitis)

Excessive alcohol intake

Chronic inflammatory conditions (systemic lupus erythematosus, rheumatoid arthritis, Sjogren’s syndrome)

 

CLASSIFICATION OF HYPERTRIGLYCERIDEMIA

 

The classification of HTG in children and adolescents, as published by the National Expert Panel on Cholesterol Levels in Children (1) and the Expert Panel on Cardiovascular Health Risk Reduction in Children (2), includes definitions of borderline and high TG based upon the 75th and 95th percentiles of TG in children, respectively. Unfortunately, this classification does not emphasize severe TG levels. Table 2 presents a classification that combines the former recommendations with the 2010 Endocrine Society guidelines on HTG (3) to focus attention on the very high levels of TG seen in primary HTG (4).

 

Hegele et al suggested a simplified classification to facilitate clinical decision-making (5). This classification uses a general population-derived distribution of plasma TG levels to define mild-to-moderate HTG if TG are 175-885 mg/dL (2-9.9 mmol/L with severe HTG defined as >885 mg/dL (10 mmol/L). The latter helps to identify those who are at increased risk of pancreatitis, who more likely to have an underlying genetic cause for HTG, and who would benefit from referred to a lipid specialist.

 

Table 2. Classification of Hypertriglyceridemia (mg/dL) in Children and Adolescents

Age

Normal

Borderline

High

Very high

Severe

Very Severe

0-9 yrs

<75

≥75-99

≥100-499

≥500-999

≥1000-1999

≥2000

10-19 yrs

<90

≥90-129

≥130-499

≥500-999

≥1000-1999

≥2000

Definitions integrated from the National Expert Panel on Blood Cholesterol Levels in Children, Expert Panel on Cardiovascular Risk Reduction in Children, and the Endocrine Society Statement on Evaluation and Treatment of Hypertriglyceridemia.

 

TRIGLYCERIDE DISORDERS IN CHILDHOOD AND ADOLESCENTS

 

Evaluation of HTG in infancy should include screening for secondary causes of HTG, particularly disorders affecting the thyroid, liver, and kidney function. Preterm and critically ill infants may be particularly prone to HTG because of immaturity, limited adipose stores, and reduced lipoprotein lipase (LPL) activity (6). In this setting, HTG may be exacerbated by stress, sepsis, selective medications, and use of intravenous fat (lipids) as a nutritional supplement. Infants with unexplained hypoglycemia and HTG should be evaluated for glycogen storage disease type I (GSD type I) especially when accompanied by hepatomegaly, lactic acidosis, and hyperuricemia.

 

Transient infantile hypertriglyceridemia (HTGTI) is an autosomal recessive hereditary disorder caused by the inactivation and variant of glycerol-3-phosphate dehydrogenase 1 located on chromosome 12q12-q13. The GPD1 gene encodes intracytoplasmic NAD-dependent GPD1, which plays an essential role in lipid and carbohydrate metabolism. In addition to HTG other manifestations include hepatomegaly, elevated liver transaminases, and hepatic steatosis in early infancy. While the HTG may normalize with age, mild HTG accompanied by elevated liver transaminases, a fatty liver, and even cirrhotic have been reported (7).

 

Genetic causes of HTG can result from rare mutations in the lipoprotein lipase (LPL) complex, where it is termed the familial chylomicronemia syndrome (FCS). More than 95% of patients with HTG have a multigenic susceptibility component, termed multifactorial chylomicronemia (MCM) (5).. Multigenic hypertriglyceridemia has a complex etiology, consisting of an excess burden of common small-effect variants, in addition to rare heterozygous large-effect variants in genes either directly or indirectly associated with plasma triglyceride concentration (8). Causes of inherited forms of severe HTG are discussed below and summarized in Table 3.

 

Table 3. Summary of Primary Hypertriglyceridemia Disorders

Lipid Disorder

Molecular Defect

Incidence

Lipoprotein Abnormality

Lipid Profile

Presentation

*Familial Chylomicronemia Syndrome (FCS)

homozygous or compound heterozygous mutations in lipoprotein lipase (LPL) *

1 per 1,000,000

↑↑ Chylomicrons,

↑↑ TG (>1000 mg/dL) and post prandial HTG > 10,000 mg/dL

Early onset ↑↑ TG, eruptive xanthomas, recurrent pancreatitis

**Familial Combined Dyslipidemia

Unknown

 

1/200

↑ VLDL,

↑ LDL

↑ TG ↑ LDL-C, ↓HDL-C, ↑ small dense LDL

Often seen with obesity, insulin resistance, hypertension

**Familial Hypertriglyceridemia

Unknown

1/500

↑↑ VLDL

↑ TG (200-1000 mg/dL)

Family members usually affected

**Dysbetalipoproteinemia

Abnormal
Apo E

1/5000

↑ Chylomicrons,
↑ VLDL remnants IDL)

↑ TG (250- 600 mg/dL); ↑ Total cholesterol

Palmer and tuberoeruptive
xanthomas

* Rare causes include mutations in apo CII, apo A-V, glycosylphosphatidylinositol-anchored high-density lipoprotein-binding protein (GPIHBP1), or lipase maturation factor-1(LMF-1) or the presence of circulating inhibitors to LPL.

**Generally present in adulthood unless precipitated by a secondary cause (obesity, insulin resistance)

 

Familial Chylomicronemia Syndrome (FCS)

 

GENETICS AND PATHOGENESIS

 

FCS has an estimated prevalence of approximately 1 in 500,000 to 1,000,000 (9, 10). FCS results from a mutation in one or more genes of the lipoprotein lipase (LPL) complex and affects catabolism of chylomicrons and very low-density lipoproteins (VLDL). The most common gene affected is LPL (accounts for 95% of these cases), in which homozygotes or compound heterozygotes inherit two defective LPL alleles. The LPL gene is composed of 10 exons and is located on chromosome 8p22. The first mutation was described in 1989, and since that time, over 100 mutations that result in LPL deficiency have been reported (11, 12). Most mutations occur in exons 3, 5, and 6, which are responsible for the catalytic coding region of the LPL gene (11). The LPL enzyme and its cofactor, apolipoprotein (apo) C-II, act on the luminal surface of the capillary endothelium and are responsible for liberating free fatty acids from the TG in dietary-derived chylomicrons and VLDL produced in the liver. When any part of the LPL complex is defective, there is a massive accumulation of chylomicrons in the blood, hence the name FCS. A lesser amount of TG from VLDL may also contribute to the observed HTG.

 

FCS may also be caused from loss of function mutations in apo C-II, the cofactor for LPL, glycosylphosphatidylinositol-anchored high density lipoprotein-binding protein (GPIHBP1), which helps to anchor chylomicrons to the endothelial surface (13), and LMF1 factor 1, an endoplasmic reticulum chaperone protein required for post-translational activation of LPL (14). Apo A-V plays a role in stabilizing the lipoprotein-enzyme complex thereby enhancing lipolysis; thus, defective or absent apo A-V can result in reduced efficiency of LPL-mediated lipolysis (15, 16). Circulating inhibitors to the LPL enzyme (17) have been described. Each of the above has an indistinguishable clinical phenotype (18).

 

PRESENTATION AND DIAGNOSIS

 

The presentation of FCS in infancy is suspected by a creamy appearance of the blood on routine blood draw or fingerstick caused from TG accumulation secondary to decreased clearance of chylomicrons from the plasma. If the diagnosis is not made from observation of a lipemic blood sample, the disease often presents as severe abdominal pain from acute pancreatitis. Recurrent abdominal pain and pancreatitis are common. The diagnosis of FCS is supported by the presence of markedly elevated TG concentrations and chylomicrons, the latter which are normally rapidly cleared from the plasma following a meal. Laboratory data will also show marked reductions in high-density lipoprotein cholesterol (HDL-C) and low-density lipoprotein cholesterol (LDL-C) concentrations (10). Homozygous or compound heterozygous individuals who have absent or markedly reduced LPL activity typically have serum TG concentrations that can reach 10,000 or higher (11). In contrast, heterozygous carriers have normal to moderately reduced LPL activity, are usually asymptomatic, and may have normal or mildly elevated fasting TG concentrations that can range from 200 to 750 mg/dL. As a result, presentation and diagnosis may occur later in childhood.

 

Physical signs may include lipemia retinalis and eruptive xanthomas, the latter generally located over the buttocks and extensor surfaces (12). Hepatosplenomegaly can occur from the accumulation of chylomicrons in the liver and spleen (12). Complications of LPL deficiency may include multiple episodes of pancreatitis leading to pancreatic calcification, diabetes mellitus, and steatorrhea, especially in those who are unable or unwilling to comply with a very low-fat diet (19).

 

Reduction or absent LPL activity can be measured after intravenous heparin administration in the presence of normal apo C-II levels (20). Heparin is a competitive agonist of LPL; absence of LPL activity after an intravenous heparin bolus is diagnostic (21). Molecular genetic analysis is also available, but is not necessary for treatment.

 

The following disorders may result in the multifactorial chylomicronemia syndrome (MFCS) that result from the co-existence of genetic and secondary forms of HTG.

 

Familial Combined Hyperlipidemia (FCHL)

 

GENETICS AND PATHOGENESIS

 

FCHL is one the most common causes of genetic hyperlipidemia with a prevalence of 0.5% to 2% in the population (22, 23). In a pediatric clinic population, FCHL has been shown to be 3 times more prevalent than familial hypercholesterolemia (24). FCHL is a genetically complex disease whose phenotype is usually determined by the interaction of multiple susceptibility genes and the environment. Genome wide association studies (GWAS) and linkage approaches have been utilized to screen the genome in FCHL families from various populations to identify loci linked to the phenotype. At least 35 genes have been implicated in the development of FCHL (polygenic disorder). One chromosomal locus that has been consistently linked to FCHL is 1q21–23 (25). Another commonly linked gene in FCHL is the ubiquitous transcription factor upstream stimulatory factor 1 (USF1), which has numerous target genes related to lipid and glucose metabolism (26). A detailed review of gene associations in FCHL is available (25). In general, the genes that have been implicated are primarily those involved in an overproduction of VLDL and apoB-100 by the liver, a reduction of fatty acid uptake by adipose tissue, and a decrease in clearance of chylomicron remnants. For those with TG 2-10 mmol/L (<885mg/dL), biochemical screening and counselling for family members is recommended, but routine genetic testing is not warranted (27).

 

PRESENTATION AND DIAGNOSIS

 

The lipid profile found in individuals with FCHL is variable. In addition to high TG concentrations, LDL-C may be normal or elevated and HDL-C levels low. There is also an increase in small dense LDL (sdLDL) particles, due to the delayed clearance of VLDL (28, 29). Elevated levels of apo B (> 90 percentile) and sdLDL particles are now considered diagnostic criteria for FCHL in adults (28), although neither is routinely assessed in clinical practice. The presence of elevated TG and apo B levels in at least 2 family members is also considered necessary for a definitive diagnosis of FCHL (30).

 

FCHL presents in childhood when unmasked by weight gain (31), and is also influenced by age (31). As a result, in normal weight individuals, the presentation can be delayed. Thus, it is possible that children with normal lipid values but a family known to have FCHL should be retested as young adults (31). Unique physical findings in FCHL are lacking, but affected individuals often exhibit obesity, signs of insulin resistance, and hypertension (32). The diagnosis is made from a characteristic fasting lipid profile and, if available, a reliable family history of dyslipidemia and early CVD (33).  

 

The association of FCHL with premature CVD is well established and CVD risk factors such as visceral adiposity, insulin resistance, impaired glucose tolerance, and hypertension are often present (34, 35). Therefore, identifying this disorder is of particular importance for management of future cardiovascular health.

 

Familial Hypertriglyceridemia (FHTG)

 

While FHTG was previously thought to be distinct from FCHL, more recent genetic characterization of individuals with familial forms of HTG indicates that both disorders are polymorphisms in multiple genes associated with HTG. Nevertheless, it is important to note that FCHL as originally described is associated with a very high prevalence of premature CVD (34-36). Thus, distinguishing clinical FCHL from FHTG may be useful for assessing cardiovascular risk.

 

PATHOGENESIS AND GENETICS

 

Familial hypertriglyceridemia (FHTG) is a polygenic disorder with a prevalence of approximately 1 per 500 (23). The genetic defect causing FHTG has not been identified, but studies in a Mexican-American cohort have identified genetic susceptibility loci on chromosomes 6, 7, and 15 that are linked to elevated TG levels (37, 38). The primary abnormality in FHTG is an overproduction of VLDL by the liver and impaired catabolism of TG-rich lipoproteins where normal numbers of very large triglyceride-enriched VLDL particles are secreted (32, 39). FHTG has also been associated with a defective regulation of bile acid synthesis, resulting in abnormally high production rate of bile acids, which associates with the subsequent development of HTG (40). Unlike FCHL, hepatic apoB-100 production is not increased and, as such, there is no overproduction of LDL. Prior work suggested that there is no increased CVD risk (23), but recent data shows baseline TG levels predicted subsequent CVD mortality after 20 years of follow up among relatives in FHTG families (34).

 

Most individuals with mild-to-moderate HTG, including those with FHTG, as well as severe elevations of TG are polygenic, with a stepwise increase in the prevalence of genetic determinants as the HTG phenotype became more severe. For individual patients, however, genetic testing alone cannot accurately predict phenotypic expression of HTG severity. At present clinical decisions regarding diet and use of lipid lowering medication should be based on the severity of the HTG, without the need for extensive genetic testing (41).

 

PRESENTATION AND DIAGNOSIS

 

TG levels are usually normal in childhood. Although FHTG is not usually expressed until adulthood, with the rise in childhood obesity FHTG has been diagnosed at an earlier age (42-44). The phenotype is usually asymptomatic HTG (42-44) with TG levels between 250 and 1000 mg/dL, normal-to-mildly elevated total cholesterol concentrations and low-to-normal LDL-C and HDL-C levels (45). The diagnosis of FHTG is made by obtaining a detailed family history and examination of fasting lipoprotein profiles of the patient and relatives.

 

Dysbetalipoproteinemia (Remnant Removal Disease)

 

PATHOGENESIS AND GENETICS

 

Dysbetalipoproteinemia is a autosomal recessive disorder with an estimated prevalence of 1 in 1000 to 1 in 2500 individuals (46). It is caused by a homozygous mutation in the apoE2 genotype (approximately 1% of population) or a dominant negative mutation in the apo E gene, which serves as a ligand for chylomicrons, intermediate-density lipoproteins, and VLDL receptors in the liver. In the presence of a secondary insult (concomitant genetic mutation, medication, or environment) there is abnormal uptake and metabolism of remnant particles (chylomicrons, intermediate-density lipoprotein, and VLDL) with subsequent accumulation of each in the blood. This disorder is an excellent example of the interaction of genetics and environment as the genetic abnormality is quite common but the expression of the clinical manifestations requires another abnormality which is frequently another disease or medication that effects lipid metabolism.

 

Isoform apoE4 is associated with an increase in LDL-cholesterol levels and thus a higher cardiovascular risk compared to apoE3; whereas apoE2 is associated with a mild decrease in LDL-cholesterol levels. Several rare APOE gene variants have been reported in different types of dyslipidemias including dysbetalipoproteinemia, familial combined hyperlipidemia (FCH), lipoprotein glomerulopathy, and autosomal dominant hypercholesterolemia due to molecular alterations in three main genes: LDLR, APOB and PCSK9. Clinical presentation of lipid disorders associated with APOE variants often strongly overlap related to common genetic and environmental factors (47).

 

PRESENTATION AND DIAGNOSIS

 

A secondary insult such as obesity, diabetes, or estrogen use is necessary for expression in childhood. The diagnosis of dysbetalipoproteinemia remnant removal disease should be suspected when total cholesterol and triglyceride levels (range from 300 to 1000 mg/dl) are roughly equal in magnitude (48). 

 

Dysbetalipoproteinemia has been documented in the pediatric age group (44, 49, 50). A case series of 3 children fromVancouver, British Columbia, Canada demonstrated early presentation of the disorder (age range, 10–11 y) due to precipitating factors including hypothyroidism, partial LPL deficiency, and concurrent familial hypercholesterolemia (50). Each child presented with palmar and tuberoeruptive xanthomas.

 

Palmer crease xanthomas (lipid deposits in the palmar creases) are pathognomonic for this condition, although eruptive xanthomas are possible on pressure sites like the elbows, knees, and buttocks (48). A 30-y retrospective review of lipid disorders from a single clinical practice identified 105 patients with dysbetalipoproteinemia. Palmar crease xanthomas occurred in 20% of patients, cutaneous xanthomas in 18%, and tendon xanthomas in 13% (48).

 

The diagnosis of dysbetalipoproteinemia is confirmed by documenting elevated remnant lipoproteins, abnormal gel electrophoresis mobility, or by identifying the genetic defect (Arg145 →Cys) in apoE2 (51). Despite having normal or low LDL cholesterol and apo B concentrations, individuals with dysbetalipoproteinemia often have an elevated CVD risk due to the increased remnant particles (52, 53). Affected individuals also are at increased risk for peripheral vascular disease (53).

 

Current lipid-based diagnostic methods have important limitations. A 3-step algorithm has been proposed for the diagnosis of dysbetalipoproteinemia using total cholesterol and TG as a first step, the non-HDL-C/apo B ratio as a second screening criterion, and finally the APOE genotype, lipoprotein ultracentrifugation, or electrophoresis as a confirmatory test (54).

 

SCREENING AND DIAGNOSIS

 

Most cases of HTG are diagnosed in childhood most often because a family member had experienced a premature cardiac event, because their siblings were known to have elevated TG levels, or because abnormal test results were obtained during a routine examination (44).

 

Screening for dyslipidemia is recommended in children ≥ 2 years who have one or more of the following: (1) parents, aunts, uncles and/or grandparents (men ≤ 55 years old, women ≤ 65 years old) who have had a heart attack, treated angina, coronary artery bypass, graft/stent/angioplasty, stroke, or sudden cardiac death; (2) parents who have high blood cholesterol levels (>240 mg/dl); or (3) parental/grandparental family history is not known, or (4) the patient has two or more other risk factors for CAD (including hypertension, cigarette smoking, low HDL cholesterol, obesity (>30% overweight), physical inactivity and diabetes mellitus (1, 55, 56). 

 

With newer recommendations of universal lipid screening between 9-11 years (2), it is likely that dysbetalipoproteinemia, and also FCHL or FHTG, may be detected more often in childhood. Any presentation of acute pancreatitis should prompt the need for a lipid profile. A fasting lipid profile (>12hours) should be obtained if TG are elevated in the non-fasting state. Cut points for normal and elevated TG levels are listed in Table 2.

 

Disorders of severe HTG are diagnosed based upon the degree of TG elevation and associated lipoprotein abnormalities (if any), the clinical features (if present), and a reliable family history, when available. Genetic testing is available for suspected cases of FCS and dysbetalipoproteinemia but is not necessary for treatment.

 

MANAGEMENT OF HTG

 

Secondary causes of HTG, including a variety of medications, are common. Therefore prior to implementation of a management plan, evaluation of secondary causes of HTG is recommended. When present, optimum treatment of secondary conditions, such as hypothyroidism, may be sufficient to correct the HTG. Medications known to cause elevations of TG should either be discontinued, if possible, or an alternative medication used.

 

Lifestyle Intervention

 

Adoption of a healthy lifestyle, including dietary modification, optimizing body weight, smoking avoidance/cessation, and physical activity, is the primary strategy for managing HTG in youth (2). Specific dietary recommendations include reducing simple carbohydrates including sugar sweetened beverages (56), substituting monounsaturated and n-6 polyunsaturated fatty acids for carbohydrate (57), and decreasing carbohydrate rich foods like white bread, rice and pasta (58). Thirty- sixty minutes of daily moderate to vigorous physical activity is also recommended for children between 2-21 years of age with TG elevations (2) as this degree of activity effectively reduces TG (59). Lifestyle recommendations for TG lowering are summarized in Table 4.

 

Table 4. Lifestyle Recommendations for Triglyceride Lowering in Children and Adolescents

1.     1) Daily caloric intake should be < 25%–30% of calories from fat, <7% from saturated fat, <200 mg/dL of cholesterol*, decrease trans fat

2.     2) Avoid sugar intake (ice cream, candy, baked goods) and sugar sweetened beverages (pop, juice, sports drinks)

3.     3) Replace simple carbohydrates (white bread, white pasta, white rice) with complex carbohydrates (wheat bread, whole grain pasta, brown rice)

4.     4) Replace carbohydrates with monounsaturated fat (olive oil, canola oil, nuts, seeds)

5.     5) Increase omega 3 fatty acids (fish)

6.     6) 30-60 minutes of moderate to vigorous exercise daily

* More severe elevations of TG may require reduction of fat to 10-15% of daily calories equivalent to ~10-25 grams/day. This is considered a very low-fat diet and should be done in consultation with a nutritionist and a pediatric lipidologist.

 

The goal of treatment in patients with FCS and MCS differs from other causes of severe HTG because these patients cannot metabolize TGs and fats. Additionally, the primary goal is prevention of pancreatitis by reducing TG concentrations in the blood. This may require a very-low-fat diet (<10-15% daily caloric intake from fat or <15-20 g/day total fat) along with restriction of simple, refined carbohydrates, though this is often difficult to maintain (60). Total carbohydrate limited to <60% daily caloric intake, adolescents advised to avoid alcohol and reproductive age females counselled about the use of oral contraceptives. Such diets should ensure 2-4% daily caloric intake of alpha linolenic and linolenic acid to meet essential fatty acid (EFA) needs.

 

As a way to increase calorie intake, medium chain triglycerides (MCTs), e.g., chain length of 10 and 12 carbons, can be considered. MCTs can be either added to infant formula or given as an oral solution to supplement fat calories. Dietary MCTs are directly absorbed into the portal vein and do not require transport by chylomicrons and as a result do not increase TG concentrations. Rouis et al. describe a unique patient with clinical features of LPL deficiency with a complete resolution of clinical symptoms with MCT oil and omega 3 fatty-acid therapy (61). It should be noted MCT oil does not contain EFAs (62).

 

Drug Treatment

 

Pharmacological management is sometimes useful in disorders resulting in severe HTG to prevent pancreatitis and/or reduce risk of CVD. Medications commonly used for TG lowering are presented in Table 5. It should be noted that although prescribed (55, 63-65), none are FDA approved for use in children and adolescents (<18 years of age) and may not be effective. In patients with FCS drug therapy with the drugs listed in table 5 are usually not effective and omega-3-fatty acids contribute to the dietary fat intake.

 

Table 5. Medications used for Triglyceride Lowering

Medication

Mechanism of Action

Lipoprotein Effects

Side Effects

Fibric Acid Derivatives*

Agonist for PPAR alpha nuclear receptors that upregulate LPL and down regulate apo C-III causing ↑degradation of VLDL and TG

↓ TG (30-60%),
↑ HDL-C

↑ LDL particle size.

Cholesterol gallstones. Contraindicated in liver and gall bladder disease.
Use caution in renal disease

Omega 3 fatty acids (fish oil) *

Decreases hepatic fatty acid and TG synthesis and VLDL release

↓ TG (20-50%),
↑ HDL-C, ↑ LDL-C,
↑ LDL particle size.

Fishy taste and burping

Nicotinic Acid*

↓ VLDL and LDL
production and HDL
degradation

↓ TG (10-40%),
↓ LDL-C, ↑ HDL-C,
↓ lipoprotein (a)

↑ LDL particle size.

Dose dependent hepatotoxicity, worsening glucose metabolism, and hyperuricemia.

*Not FDA approved for <18 years of age

 

Fibric acid derivatives (fenofibrate, gemfibrozil) lower blood TG levels by reducing VLDL production and promoting catabolism of TG through enhanced LPL activity. In general, fibrates lower TG concentrations by 30-60% (66, 67). They have a modest effect on increasing HDL-C levels but can increase LDL particle size. Since FCS results from a lack of LPL activity, a response to fibrates is not expected in FCS. Fibrates are usually effective in MCS as there is typically some LPL activity. Rare side effects include dyspepsia, diarrhea, an increase in transaminases, cholelithiasis, and deep venous thrombosis. Fibrates must be used with caution in patients with renal dysfunction and gall bladder disease. In most cases, fenofibrate is used, but gemfibrozil is preferred in renal insufficiency.

 

Long chain omega 3 fatty acids inhibit diacyl glycerol acetyl transferase (DGAT), reduce VLDL-TG synthesis, and increase the rate of peroxisomal beta oxidation in the liver. In adults, omega 3 fatty acids lower TG levels by 20-50% (68). These effects are primarily seen with prescription fish oils which contain approximately 465 mg of eicosapentaenoic acid (EPA) and 375 mg of docosahexaenoic acid (DHA) and require taking at least 2 grams of omega 3 fatty acids per day. Over the counter preparations have variable quantities of EPA and DHA resulting in variable TG lowering effects. Two small studies in children with hypertriglyceridemia did not find significant lowering of the TG levels (69, 70), however, the studies were likely underpowered. When using omega- 3 fatty acids in patients with severe hypertriglyceridemia it is important to ensure that the total fat intake is within the fat allowance permitted. Four capsules of omega-3 fatty acids provide 4 grams of fat/day.

 

Niacin lowers TGs 10-30%, increases HDL cholesterol by 10-40% and lowers LDL cholesterol by 5-20%. The most common complaint with its use is flushing due to the release of prostaglandin E2 in the skin. Flushing typically occurs 15-60 minutes after ingestion and can last up to 30 minutes. Aspirin 30 minutes before niacin can reduce flushing. Children often do not tolerate niacin.

 

Patients with LPL deficiency can be offered a trial of fibric acid derivatives but the response is quite variable since these agents work to lower plasma TG primarily by upregulating LPL activity, which is deficient in this condition (71). Omega 3 fatty acids lower plasma TGs in certain conditions of HTG, but they may actually aggravate the severe HTG of FCS and are therefore contraindicated in LPL deficiency (61).

 

Volanesorsen is an antisense oligonucleotide (ASO) which binds to and induces degradation of APO C3 mRNA in the hepatocyte, resulting in reduced apo C-III protein synthesis. The drug is administered by sub-cutaneous injection and mostly cleared through the kidney. Short term clinical trials demonstrated improved lipid profiles following weekly Volanesorsen injection in patients with severe HTG due to heterogeneous causes of HTG.

 

APPROACH, a phase 3, double-blind clinical trial randomized 66 patients with familial chylomicronemia syndrome to Volanesorsen or placebo for 52-week to evaluate the safety and effectiveness. Patients receiving Volanesorsen had a 77% decrease in mean TG levels, corresponding to a mean decrease of 1,712 mg/dL (19.3 mmol/L). TG levels less than 750 mg/dL were achieved in 77% of patients with FCS. Thrombocytopenia and injection-site reactions were common adverse events (72).

 

The COMPASS trial was a randomized, placebo-controlled, double-blind, phase 3 study done at 38 international clinical sites. Subjects were 18 years-of-age or older with multifactorial severe HTG or FCS, who had a BMI of <45 kg/m2 and fasting plasma TG >500 mg/dL. Subjects were randomly assigned to subcutaneous Volanesorsen or placebo once a week for 26 weeks. Volanesorsen reduced mean plasma TG concentration by 71.2% from baseline to 3 months compared with the placebo group, representing a mean absolute reduction of fasting plasma TG of 869 mg/dL. The most common adverse event were injection-site reactions (average of 24% of all Volanesorsen injections vs 0.2% of placebo injections), which were all mild or moderate. One participant in the Volanesorsen group had thrombocytopenia and one patient experienced serum sickness (73). Rejected in 2018 by the U.S. FDA due to adverse effects on platelets, the EMA approved Volanesorsen for treatment of FSC in Europe the following year.  

 

Acute management of HTG requires maintaining NPO status, especially if there is concomitant pancreatitis. A short-term insulin infusion can be tried especially in patients with diabetes, as insulin enhances LPL activity. An intravenous infusion of regular insulin at a rate of 0.1 to 0.3 units/kg/hour while monitoring blood glucose levels will result in a reduction of TG levels down by 40-80% in 24-48 hours (74-78). TG levels can be measured every 12-24 hours during insulin infusion, and glucose levels should be monitored every hour. 

 

In individuals with primary HTG who have continued pancreatitis, plasmapheresis has been utilized. In this procedure plasma is separated from the blood and processed to eliminate selective components. The plasma is then reinfused, though on occasions it may be completely eliminated and replaced by an isovolumetric solution. Plasmapheresis can be carried out as either an emergency or a scheduled procedure. In situations where urgent, rapid and efficient reduction in TG levels is needed, such as in pancreatitis, plasmapheresis has proven a valid and safe technique and results in reductions of TG as much as 60% (79). A multicenter study recently published data demonstrating success using plasmapheresis to prevent pancreatitis in those who fail medical therapy (80).

 

Novel Therapies

 

Novel therapies for treatment of HTG are in development (Table 6). These agents increase clearance or reduce the production of triglyceride rich lipoproteins. Their clinical efficacy, cost-effectiveness, and indications, especially in children, have not yet been established.

 

Table 6. Triglyceride Lowering Agents in Development

Mechanism of action

Class/Drug

TG Lowering

Predominant Side Effects

Decreased production of TG/TRLP

MTP inhibitor (lomitapide)

35-65%

Mild GI side effects, transaminitis

 

Apo B ASO

(mipomersen)

8-10%

Injection site reactions, flu like symptoms and transaminitis

Increased TG/TRLP catabolism

ANGPTL3 mAb (evinacumab)

77-83%

Flu-like symptoms, dizziness, myalgia, nausea

 

ANGPTL3 ASO (vupanorsen)

40-50%

Injection site reactions

 

 

COMPLICATIONS OF HTG

 

Cardiovascular Disease

 

Children and adolescents with persistently moderate to high levels of TG may be at increased risk for premature cardiovascular disease during adulthood. However, the extent to which HTG independently contributes to CVD has long been debated and remains unknown (3, 81-83). Some studies have shown an independent relationship between HTG and CVD, but effect sizes are small but significant when adjusted for both HDL and non-HDL-C (84, 85).

 

In FCHL the increased CVD risk in probands and first degree relatives is largely attributed to the increase in apo B (34) and/or lipoprotein (a) (86). Likewise, in dysbetalipoproteinemia the increased CVD risk is attributed to increased remnant lipoprotein particles (52, 53). A recent systematic review and meta-analysis of observational studies evaluating HTG and CVD found that fasting HTG was associated with an increase in cardiovascular death (odds ratios (OR) 1.80; 95% confidence interval (CI) 1.31-2.49), cardiovascular events (OR, 1.37; 95% CI, 1.23-1.53), and myocardial infarction (OR, 1.31; 95% CI, 1.15-1.4 (87).

 

To address a causal role for TG in the development of CVD, several Medellin randomization studies have been conducted. While TG raising variant alleles have been associated with clinical CVD endpoints, in most cases, a second lipid disturbance—usually depressed HDL-C—was concurrently associated (88). It appears TG may be part of a joint phenotype or a biomarker of metabolic risk that leads to CVD.

 

Pemafibrate, a selective peroxisome proliferator-activated receptor α modulator, has previously been shown to reduce TG levels and improves other lipid levels. In a multinational, double-blind, randomized, controlled trial of patients with type 2 diabetes, mild-to-moderate HTG (TG level, 200 to 499 mg/dL) and low HDL (HDL-C <40 mg/dL) Pemafibrate lowered TG, VLDL cholesterol, remnant cholesterol, and apolipoprotein C-III levels. However, the incidence of cardiovascular events was not lower among those who received Pemafibrate than among those who received placebo (89). In a separate trial, Pemafibrate did not decrease liver fat content but had significant reduction in magnetic resonance imaging-estimated proton density fat fraction-based liver stiffness (90).

 

Pancreatitis

 

FCS commonly presents with spontaneous pancreatitis in the first decade of life as a result of the degree of TG elevation. In contrast, FHTG and dysbetalipoproteinemia usually require a secondary risk factor to incite pancreatitis in adolescence (42, 50).

 

HTG accounts for 1-4% of cases of acute pancreatitis (91). Though the exact mechanism of inciting pancreatitis is unknown, TG-rich chylomicrons are thought to impair circulatory flow in capillary beds of the pancreas causing ischemia and triggering an inflammatory response (11, 12). HTG is the most common cause of pancreatitis not due to gallstones or alcohol abuse (1, 2, 55, 92).  

 

Pancreatitis generally occurs when TG levels exceed 1000-1500 mg/dL (93, 94) but TG between 200-1000 mg/dL can be seen in the early stages of acute pancreatitis of any etiology (95, 96). The risk of developing acute pancreatitis with serum TG >1000 and >2000 mg/dL is 5% and 10% to 20%, respectively (94). The presentation of pancreatitis usually includes abdominal pain, vomiting, and ileus (97). When the diagnosis is suspected, serum TG levels should be measured since elevated concentrations in the blood can diminish rapidly. Thus, a delay in obtaining TG concentration may lead to falsely low levels. Prevention of pancreatitis relies on consistent TG lowering. Lowering levels to < 500 mg/dL effectively prevents recurrences of pancreatitis in most affected individuals (94). Prevention of pancreatitis is crucial since mortality from pancreatitis can be as high as 20% (98).  

 

CONCLUSIONS

 

Identification of genetic causes of severe HTG in pediatric patients is important given the risk for pancreatitis and/or early CVD. Lifestyle modification is central to prevention, but often is not sufficient. While medications can be helpful in lowering TG, in some disorders they have no benefit. Novel therapies may be on the horizon. Whether these therapies will be beneficial in treating primary disorders of HTG in children and adolescents and their associated complications remains to be seen.

 

REFERENCES

 

  1. National Cholesterol Education Program (NCEP): highlights of the report of the Expert Panel on Blood Cholesterol Levels in Children and Adolescents. Pediatrics. Mar 1992;89(3):495-501.
  2. Expert panel on integrated guidelines for cardiovascular health and risk reduction in children and adolescents: summary report. Pediatrics. Dec 2011;128 Suppl 5:S213-56. doi:10.1542/peds.2009-2107C
  3. Berglund, L., et al. Evaluation and treatment of hypertriglyceridemia: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. Sep 2012;97(9):2969-89. doi:10.1210/jc.2011-3213
  4. Shah, A.S. and D.P. Wilson. Primary hypertriglyceridemia in children and adolescents. J Clin Lipidol. Sep-Oct 2015;9(5 Suppl):S20-8. doi:10.1016/j.jacl.2015.04.004
  5. Dron, J.S. and R.A. Hegele. Genetics of Hypertriglyceridemia. Front Endocrinol (Lausanne). 2020;11:455. doi:10.3389/fendo.2020.00455
  6. Shennan, A.T., M.H. Bryan, and A. Angel. The effect of gestational age on intralipid tolerance in newborn infants. J Pediatr. Jul 1977;91(1):134-7. doi:10.1016/s0022-3476(77)80465-4
  7. Wang, J., et al. Clinical characteristics and variant analyses of transient infantile hypertriglyceridemia related to GPD1 gene. Front Genet. 2022;13:916672. doi:10.3389/fgene.2022.916672
  8. Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34). UK Prospective Diabetes Study (UKPDS) Group. Lancet. Sep 12 1998;352(9131):854-65.
  9. Hayden, M.R. and Y. Ma. Molecular genetics of human lipoprotein lipase deficiency. Mol Cell Biochem. Aug 18 1992;113(2):171-6.
  10. Santamarina-Fojo, S. The familial chylomicronemia syndrome. Endocrinol Metab Clin North Am. Sep 1998;27(3):551-67, viii.
  11. Gilbert, B., et al. Lipoprotein lipase (LPL) deficiency: a new patient homozygote for the preponderant mutation Gly188Glu in the human LPL gene and review of reported mutations: 75 % are clustered in exons 5 and 6. Ann Genet. Jan-Mar 2001;44(1):25-32.
  12. Rahalkar, A.R., et al. Novel LPL mutations associated with lipoprotein lipase deficiency: two case reports and a literature review. Can J Physiol Pharmacol. Mar 2009;87(3):151-60. doi:10.1139/y09-005
  13. Johansen, C.T. and R.A. Hegele. Genetic bases of hypertriglyceridemic phenotypes. Curr Opin Lipidol. Aug 2011;22(4):247-53. doi:10.1097/MOL.0b013e3283471972
  14. Peterfy, M. Lipase maturation factor 1: a lipase chaperone involved in lipid metabolism. Biochim Biophys Acta. May 2012;1821(5):790-4. doi:10.1016/j.bbalip.2011.10.006
  15. Priore Oliva, C., et al. Inherited apolipoprotein A-V deficiency in severe hypertriglyceridemia. Arterioscler Thromb Vasc Biol. Feb 2005;25(2):411-7. doi:10.1161/01.ATV.0000153087.36428.dd
  16. Nilsson, S.K., et al. Apolipoprotein A-V; a potent triglyceride reducer. Atherosclerosis. Nov 2011;219(1):15-21. doi:10.1016/j.atherosclerosis.2011.07.019
  17. Brunzell, J.D., et al. Familial chylomicronemia due to a circulating inhibitor of lipoprotein lipase activity. J Lipid Res. Jan 1983;24(1):12-9.
  18. Fredrickson, D.S. and R.S. Lees. A SYSTEM FOR PHENOTYPING HYPERLIPOPROTEINEMIA. Circulation. Mar 1965;31:321-7.
  19. Kawashiri, M.A., et al. Long-term course of lipoprotein lipase (LPL) deficiency due to homozygous LPL(Arita) in a patient with recurrent pancreatitis, retained glucose tolerance, and atherosclerosis. J Clin Endocrinol Metab. Dec 2005;90(12):6541-4. doi:10.1210/jc.2005-1098
  20. Fojo, S.S. and H.B. Brewer. Hypertriglyceridaemia due to genetic defects in lipoprotein lipase and apolipoprotein C-II. J Intern Med. Jun 1992;231(6):669-77.
  21. Eckel, R.H. Lipoprotein lipase. A multifunctional enzyme relevant to common metabolic diseases. N Engl J Med. Apr 20 1989;320(16):1060-8. doi:10.1056/nejm198904203201607
  22. Holmes, K.W. and P.O. Kwiterovich, Jr. Treatment of dyslipidemia in children and adolescents. Curr Cardiol Rep. Nov 2005;7(6):445-56.
  23. Brunzell, J.D. Clinical practice. Hypertriglyceridemia. N Engl J Med. Sep 6 2007;357(10):1009-17. doi:10.1056/NEJMcp070061
  24. Cortner, J.A., P.M. Coates, and P.R. Gallagher. Prevalence and expression of familial combined hyperlipidemia in childhood. J Pediatr. Apr 1990;116(4):514-9.
  25. Brouwers, M.C., et al. The genetics of familial combined hyperlipidaemia. Nat Rev Endocrinol. Jun 2012;8(6):352-62. doi:10.1038/nrendo.2012.15
  26. Suviolahti, E., H.E. Lilja, and P. Pajukanta. Unraveling the complex genetics of familial combined hyperlipidemia. Ann Med. 2006;38(5):337-51. doi:10.1080/07853890600865759
  27. Hegele, R.A., et al. The polygenic nature of hypertriglyceridaemia: implications for definition, diagnosis, and management. Lancet Diabetes Endocrinol. Aug 2014;2(8):655-66. doi:10.1016/s2213-8587(13)70191-8
  28. Ayyobi, A.F., et al. Small, dense LDL and elevated apolipoprotein B are the common characteristics for the three major lipid phenotypes of familial combined hyperlipidemia. Arterioscler Thromb Vasc Biol. Jul 1 2003;23(7):1289-94. doi:10.1161/01.atv.0000077220.44620.9b
  29. Pauciullo, P., et al. Small dense low-density lipoprotein in familial combined hyperlipidemia: Independent of metabolic syndrome and related to history of cardiovascular events. Atherosclerosis. Mar 2009;203(1):320-4. doi:10.1016/j.atherosclerosis.2008.07.004
  30. Sniderman, A., S.D. Bailey, and J.C. Engert. Familial combined hyperlipidaemia: how can genetic disorders be common, complex and comprehensible? Clin Sci (Lond). Nov 2007;113(9):365-7. doi:10.1042/cs20070220
  31. Shamir, R., et al. The influence of age and relative weight on the presentation of familial combined hyperlipidemia in childhood. Atherosclerosis. Mar 1996;121(1):85-91.
  32. Durrington, P. Dyslipidaemia. Lancet. Aug 30 2003;362(9385):717-31. doi:10.1016/s0140-6736(03)14234-1
  33. Goldstein, J.L., et al. Hyperlipidemia in coronary heart disease. II. Genetic analysis of lipid levels in 176 families and delineation of a new inherited disorder, combined hyperlipidemia. J Clin Invest. Jul 1973;52(7):1544-68. doi:10.1172/jci107332
  34. Austin, M.A., et al. Cardiovascular disease mortality in familial forms of hypertriglyceridemia: A 20-year prospective study. Circulation. Jun 20 2000;101(24):2777-82.
  35. Brunzell, J.D., et al. Myocardial infarction in the familial forms of hypertriglyceridemia. Metabolism. Mar 1976;25(3):313-20. doi:10.1016/0026-0495(76)90089-5
  36. McNeely, M.J., et al. Lipoprotein and apolipoprotein abnormalities in familial combined hyperlipidemia: a 20-year prospective study. Atherosclerosis. Dec 2001;159(2):471-81. doi:10.1016/s0021-9150(01)00528-7
  37. Arya, R., et al. Factors of insulin resistance syndrome--related phenotypes are linked to genetic locations on chromosomes 6 and 7 in nondiabetic mexican-americans. Diabetes. Mar 2002;51(3):841-7.
  38. Duggirala, R., et al. A major susceptibility locus influencing plasma triglyceride concentrations is located on chromosome 15q in Mexican Americans. Am J Hum Genet. Apr 2000;66(4):1237-45. doi:10.1086/302849
  39. Chait, A., J.J. Albers, and J.D. Brunzell. Very low density lipoprotein overproduction in genetic forms of hypertriglyceridaemia. Eur J Clin Invest. Feb 1980;10(1):17-22.
  40. Angelin, B., K.S. Hershon, and J.D. Brunzell. Bile acid metabolism in hereditary forms of hypertriglyceridemia: evidence for an increased synthesis rate in monogenic familial hypertriglyceridemia. Proc Natl Acad Sci U S A. Aug 1987;84(15):5434-8.
  41. Dron, J.S., et al. Severe hypertriglyceridemia is primarily polygenic. J Clin Lipidol. Jan-Feb 2019;13(1):80-88. doi:10.1016/j.jacl.2018.10.006
  42. Bouaziz Abed, A., et al. [Primary hypertriglyceridemia type IV : early presentation of a severe case]. Tunis Med. Nov 2013;91(11):683. Hypertriglyceridemie primaire de type iv=forme precoce et severe de decouverte fortuite.
  43. Clauss, S.B. and P.O. Kwiterovich Jr. Genetic disorders of lipoprotein transport in children. Progress in Pediatric Cardiology. 9// 2003;17(2):123-133. doi:http://dx.doi.org/10.1016/S1058-9813(03)00049-3
  44. Manlhiot, C., et al. Spectrum and management of hypertriglyceridemia among children in clinical practice. Pediatrics. Feb 2009;123(2):458-65. doi:10.1542/peds.2008-0367
  45. Tullu, M.S., et al. Familial hypertriglyceridemia. Indian J Pediatr. Dec 2008;75(12):1257-8. doi:10.1007/s12098-008-0219-y
  46. Smelt, A.H. and F. de Beer. Apolipoprotein E and familial dysbetalipoproteinemia: clinical, biochemical, and genetic aspects. Semin Vasc Med. Aug 2004;4(3):249-57. doi:10.1055/s-2004-861492
  47. Khalil, Y.A., et al. APOE gene variants in primary dyslipidemia. Atherosclerosis. Jul 2021;328:11-22. doi:10.1016/j.atherosclerosis.2021.05.007
  48. Blom, D.J., et al. Dysbetalipoproteinaemia--clinical and pathophysiological features. S Afr Med J. Nov 2002;92(11):892-7.
  49. Ziemer, A. and H.D. Goring. [Severe type III hyperlipoproteinemia with unusual lipoprotein phenotype in an adolescent patient]. Hautarzt. Aug 1993;44(8):538-44. Schwere Typ-III-Hyperlipoproteinaimie mit ungewohnlichem Lipoproteinphanotyp bei einer jugendlichen Patientin.
  50. Fung, M., et al. Case series of type III hyperlipoproteinemia in children. BMJ Case Rep. 2011;2011doi:10.1136/bcr.02.2011.3895
  51. Marais, A.D., G.A. Solomon, and D.J. Blom. Dysbetalipoproteinaemia: a mixed hyperlipidaemia of remnant lipoproteins due to mutations in apolipoprotein E. Crit Rev Clin Lab Sci. Feb 2014;51(1):46-62. doi:10.3109/10408363.2013.870526
  52. Mahley, R.W. Apolipoprotein E: cholesterol transport protein with expanding role in cell biology. Science. Apr 29 1988;240(4852):622-30.
  53. Mahley, R.W., et al. Lipoproteins of special significance in atherosclerosis. Insights provided by studies of type III hyperlipoproteinemia. Ann N Y Acad Sci. 1985;454:209-21.
  54. Paquette, M., et al. A simplified diagnosis algorithm for dysbetalipoproteinemia. J Clin Lipidol. Jul-Aug 2020;14(4):431-437. doi:10.1016/j.jacl.2020.06.004
  55. Christian, J.B., et al. Prevalence, characteristics, and risk factors of elevated triglyceride levels in US children. Clin Pediatr (Phila). Dec 2011;50(12):1103-9. doi:10.1177/0009922811414286
  56. Johnson, R.K., et al. Dietary sugars intake and cardiovascular health: a scientific statement from the American Heart Association. Circulation. Sep 15 2009;120(11):1011-20. doi:10.1161/circulationaha.109.192627
  57. Mensink, R.P., et al. Effects of dietary fatty acids and carbohydrates on the ratio of serum total to HDL cholesterol and on serum lipids and apolipoproteins: a meta-analysis of 60 controlled trials. Am J Clin Nutr. May 2003;77(5):1146-55.
  58. Wolever, T.M., et al. The glycemic index: methodology and clinical implications. Am J Clin Nutr. Nov 1991;54(5):846-54.
  59. Graham, T.E. Exercise, postprandial triacylglyceridemia, and cardiovascular disease risk. Can J Appl Physiol. Dec 2004;29(6):781-99.
  60. Yuan, G., K.Z. Al-Shali, and R.A. Hegele. Hypertriglyceridemia: its etiology, effects and treatment. CMAJ. Apr 10 2007;176(8):1113-20. doi:10.1503/cmaj.060963
  61. Rouis, M., et al. Therapeutic response to medium-chain triglycerides and omega-3 fatty acids in a patient with the familial chylomicronemia syndrome. Arterioscler Thromb Vasc Biol. Jul 1997;17(7):1400-6.
  62. Williams, L., et al. Nutrition interventions for youth with dyslipidemia: a National Lipid Association clinical perspective. J Clin Lipidol. Nov-Dec 2022;16(6):776-796. doi:10.1016/j.jacl.2022.07.011
  63. Araujo, R.S., P. Ramos Ade, and A. Borges Mde. [Bezafibrate in an infant with congenital generalized lipodystrophy and severe hypertriglyceridemia]. Arq Bras Endocrinol Metabol. Nov 2013;57(8):653-8. Bezafibrato em lactente portador de lipodistrofia generalizada congenita e hipertrigliceridemia grave.
  64. Ellis, D., et al. Atypical hyperlipidemia and nephropathy associated with apolipoprotein E homozygosity. J Am Soc Nephrol. Oct 1995;6(4):1170-7.
  65. Vivatrat, N., B.A. Barshop, and K.L. Jones. Severe hypertriglyceridemia and recurrent pancreatitis in a girl with type Ia glycogen storage disease and type III hyperlipoproteinemia. Am J Med Genet A. Nov 2009;149A(11):2557-9. doi:10.1002/ajmg.a.33046
  66. Frick, M.H., et al. Helsinki Heart Study: primary-prevention trial with gemfibrozil in middle-aged men with dyslipidemia. Safety of treatment, changes in risk factors, and incidence of coronary heart disease. N Engl J Med. Nov 12 1987;317(20):1237-45. doi:10.1056/nejm198711123172001
  67. Keech, A., et al. Effects of long-term fenofibrate therapy on cardiovascular events in 9795 people with type 2 diabetes mellitus (the FIELD study): randomised controlled trial. Lancet. Nov 26 2005;366(9500):1849-61. doi:10.1016/s0140-6736(05)67667-2
  68. Musa-Veloso, K., et al. Long-chain omega-3 fatty acids eicosapentaenoic acid and docosahexaenoic acid dose-dependently reduce fasting serum triglycerides. Nutr Rev. Mar 2010;68(3):155-67. doi:10.1111/j.1753-4887.2010.00272.x
  69. de Ferranti, S.D., et al. Using high-dose omega-3 fatty acid supplements to lower triglyceride levels in 10- to 19-year-olds. Clin Pediatr (Phila). May 2014;53(5):428-38. doi:10.1177/0009922814528032
  70. Gidding, S.S., et al. A double-blind randomized trial of fish oil to lower triglycerides and improve cardiometabolic risk in adolescents. J Pediatr. Sep 2014;165(3):497-503.e2. doi:10.1016/j.jpeds.2014.05.039
  71. Grundy, S.M. and G.L. Vega. Fibric acids: effects on lipids and lipoprotein metabolism. Am J Med. Nov 27 1987;83(5B):9-20.
  72. Witztum, J.L., et al. Volanesorsen and Triglyceride Levels in Familial Chylomicronemia Syndrome. N Engl J Med. Aug 8 2019;381(6):531-542. doi:10.1056/NEJMoa1715944
  73. Gouni-Berthold, I., et al. Efficacy and safety of volanesorsen in patients with multifactorial chylomicronaemia (COMPASS): a multicentre, double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Diabetes Endocrinol. May 2021;9(5):264-275. doi:10.1016/s2213-8587(21)00046-2
  74. Jabbar, M.A., M.I. Zuhri-Yafi, and J. Larrea. Insulin therapy for a non-diabetic patient with severe hypertriglyceridemia. J Am Coll Nutr. Oct 1998;17(5):458-61. doi:10.1080/07315724.1998.10718794
  75. Mikhail, N., et al. Treatment of severe hypertriglyceridemia in nondiabetic patients with insulin. Am J Emerg Med. May 2005;23(3):415-7. doi:10.1016/j.ajem.2005.02.036
  76. Ippisch, H.M., et al. Hypertriglyceridemia Induced Pancreatitis: Inpatient Management at a Single Pediatric Institution. Pancreas. 2020;49(3):429-434.
  77. Lawson, E.B., M. Gottschalk, and D.E. Schiff. Insulin infusion to treat severe hypertriglyceridemia associated with pegaspargase therapy: a case report. Journal of pediatric hematology/oncology. 2011;33(2):e83-e86.
  78. Rodríguez Santana, Y., et al. Treatment of severe hypertriglyceridemia with continuous insulin infusion. Case reports in critical care. 2011;2011
  79. Ewald, N. and H.U. Kloer. Severe hypertriglyceridemia: an indication for apheresis? Atheroscler Suppl. Dec 29 2009;10(5):49-52. doi:10.1016/s1567-5688(09)71810-0
  80. Stefanutti, C., et al. Therapeutic plasma exchange in patients with severe hypertriglyceridemia: a multicenter study. Artif Organs. Dec 2009;33(12):1096-102.
  81. Hulley, S.B., et al. Epidemiology as a guide to clinical decisions. The association between triglyceride and coronary heart disease. N Engl J Med. Jun 19 1980;302(25):1383-9. doi:10.1056/nejm198006193022503
  82. Hokanson, J.E. and M.A. Austin. Plasma triglyceride level is a risk factor for cardiovascular disease independent of high-density lipoprotein cholesterol level: a meta-analysis of population-based prospective studies. J Cardiovasc Risk. Apr 1996;3(2):213-9.
  83. Miller, M., et al. Triglycerides and cardiovascular disease: a scientific statement from the American Heart Association. Circulation. May 24 2011;123(20):2292-333. doi:10.1161/CIR.0b013e3182160726
  84. Criqui, M.H., et al. Plasma triglyceride level and mortality from coronary heart disease. N Engl J Med. Apr 29 1993;328(17):1220-5. doi:10.1056/nejm199304293281702
  85. Fontbonne, A., et al. Hypertriglyceridaemia as a risk factor of coronary heart disease mortality in subjects with impaired glucose tolerance or diabetes. Results from the 11-year follow-up of the Paris Prospective Study. Diabetologia. May 1989;32(5):300-4.
  86. Kim, H., et al. Lipoprotein(a) as a risk factor for maternal cardiovascular disease mortality in kindreds with familial combined hyperlipidemia or familial hypertriglyceridemia. Clin Genet. Sep 2001;60(3):188-97.
  87. Murad, M.H., et al. The association of hypertriglyceridemia with cardiovascular events and pancreatitis: a systematic review and meta-analysis. BMC Endocr Disord. 2012;12:2. doi:10.1186/1472-6823-12-2
  88. Dron, J.S. and R.A. Hegele. Genetics of Triglycerides and the Risk of Atherosclerosis. Curr Atheroscler Rep. Jul 2017;19(7):31. doi:10.1007/s11883-017-0667-9
  89. Das Pradhan, A., et al. Triglyceride Lowering with Pemafibrate to Reduce Cardiovascular Risk. N Engl J Med. Nov 24 2022;387(21):1923-1934. doi:10.1056/NEJMoa2210645
  90. Sasaki, Y., et al. Pemafibrate, a selective PPARα modulator, prevents non-alcoholic steatohepatitis development without reducing the hepatic triglyceride content. Sci Rep. May 8 2020;10(1):7818. doi:10.1038/s41598-020-64902-8
  91. Fortson, M.R., S.N. Freedman, and P.D. Webster, 3rd. Clinical assessment of hyperlipidemic pancreatitis. Am J Gastroenterol. Dec 1995;90(12):2134-9.
  92. Executive Summary of The Third Report of The National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, And Treatment of High Blood Cholesterol In Adults (Adult Treatment Panel III). JAMA. May 16 2001;285(19):2486-97.
  93. Athyros, V.G., et al. Long-term follow-up of patients with acute hypertriglyceridemia-induced pancreatitis. J Clin Gastroenterol. Apr 2002;34(4):472-5.
  94. Scherer, J., et al. Issues in hypertriglyceridemic pancreatitis: an update. J Clin Gastroenterol. Mar 2014;48(3):195-203. doi:10.1097/01.mcg.0000436438.60145.5a
  95. Balanescu, N.R., et al. Acute pancreatitis secondary to hyperlipidemia in an 11-year-old girl: a case report and review of literature. J Med Life. Mar 15 2013;6(1):2-6.
  96. Dominguez-Munoz, J.E., et al. Hyperlipidemia in acute pancreatitis. Relationship with etiology, onset, and severity of the disease. Int J Pancreatol. Nov-Dec 1991;10(3-4):261-7.
  97. Sanchez-Ramirez, C.A., et al. Acute and recurrent pancreatitis in children: etiological factors. Acta Paediatr. Apr 2007;96(4):534-7. doi:10.1111/j.1651-2227.2007.00225.x
  98. Gan, S.I., et al. Hypertriglyceridemia-induced pancreatitis: A case-based review. World J Gastroenterol. Nov 28 2006;12(44):7197-202.

Approach to the Patient with Dyslipidemia

ABSTRACT

 

In evaluating a patient with dyslipidemia the initial step is to decide which particular lipid/lipoprotein abnormalities need to be evaluated and whether they need treatment. These disorders can be divided into elevations of LDL-C, triglycerides, non-HDL-C, and Lp(a) and decreases in plasma HDL-C. Frequently a patient can have multiple lipid/lipoprotein abnormalities. The next step is to rule out secondary causes that could account for the abnormal lipid/lipoprotein levels. These secondary causes can be due to diet, various disease states, or drug therapy. One should be suspicious of a secondary cause if a patient suddenly develops a lipid/lipoprotein abnormality or the lipid/lipoprotein profile suddenly worsens. Next one should consider the possibility of a genetic disorder and therefore ask whether relatives have either premature cardiovascular disease, lipid disorders, or are receiving lipid lowering medications. If the triglyceride levels are markedly elevated one should inquire about a family history of pancreatitis. When the lipid/lipoprotein abnormality is markedly abnormal or begins at a young age, the likelihood of a genetic disorder is increased and the family history assumes even greater importance. In most circumstances a routine lipid panel consisting of plasma triglycerides, total cholesterol, HDL-C, and calculated LDL-C and non-HDL-C provides sufficient information to appropriately decide on who to treat and the best treatment approach. However, it should be recognized that there are certain situations where more sophisticated and detailed laboratory studies can be helpful. The purpose of treating lipid disorders is to prevent the development of other diseases, particularly cardiovascular disease. Thus, the decision to treat should be based on the risk of the hyperlipidemia leading to those medical problems. Several guidelines have been published that discuss in detail cardiovascular risk assessment and provide recommendations on treatment strategies. Additionally, calculators are available on-line to determine an individual patient’s risk of developing cardiovascular disease in the next 10 years or their lifetime risk. In the prevention of cardiovascular disease, the main priority is to lower the LDL-C levels. Reductions in other apolipoprotein B containing lipoproteins may be instituted if LDL-C levels are at goal. Depending on the specific guideline the percent reduction in LDL-C and/or the goal LDL-C will vary depending upon the patient profile. When LDL-C levels are at goal but triglyceride and non-HDL-C levels are still elevated a recent study suggests further treatment with icosapent ethyl may be beneficial. Whether decreasing Lp(a) is beneficial in preventing cardiovascular disease is uncertain and further studies are in progress. Lifestyle changes are the initial treatment but in most patients’ drug therapy will be necessary.

 

INTRODUCTION

 

The initial step is to decide which particular lipid/lipoprotein abnormalities need to be evaluated and whether they need treatment. These disorders can be divided into elevations of LDL-C, triglycerides, non-HDL-C, and Lp(a), and decreases in HDL-C. An increase in non-HDL-C accompanies an increase in LDL-C and/or triglycerides levels. Often a patient can have multiple lipid/lipoprotein abnormalities. For example, it is not uncommon for a patient to have high triglycerides with low HDL-C levels or high LDL-C and high Lp(a) levels.

 

From a clinical point of view, one is not usually concerned if the LDL-C, Lp(a), or triglyceride levels are low or if the HDL-C level is high. Very low levels of LDL-C and/or triglycerides suggest the presence of other medical issues such as hyperthyroidism, malabsorption, liver disease, chronic infections, cancer, etc. On rare occasions very low LDL-C levels or triglyceride levels can be due to genetic disorders (1). Very high HDL-C levels can also be due to genetic causes (2).

 

RULE OUT SECONDARY CAUSES

 

The next step is to rule out secondary causes that could account for the abnormal lipid/lipoprotein levels. These secondary causes can be due to diet, various disease states, or medications. One should be suspicious of a secondary cause if a patient suddenly develops a lipid/lipoprotein abnormality or the lipid/lipoprotein profile suddenly worsens. Patients with genetic abnormalities causing dyslipidemia can have their disorder worsen if they develop secondary causes that further adversely affect lipid/lipoprotein levels.

 

The key is that if one corrects the secondary cause the lipid/lipoprotein abnormality can often markedly improve or even disappear. For example, hypothyroidism can be accompanied by striking increases in LDL-C levels and the treatment of hypothyroidism can result in a large decrease in LDL-C, often to normal levels (3). Likewise, an improvement in glycemic control in a patient with poorly controlled diabetes may result in a large decrease in serum triglyceride levels (4). Occasionally, the presence of dyslipidemia leads to the discovery of an unrecognized secondary disorder that requires treatment.

 

Similarly stopping certain drugs can greatly improve the lipid profile (5). For example, in some postmenopausal women with hypertriglyceridemia stopping oral estrogen therapy can result in a marked decrease in triglyceride levels (3). The disorders and drugs that cause lipid/lipoprotein abnormalities are shown in tables 1-7. It should be noted that many disorders and drugs can cause multiple lipid abnormalities. The effects of disorders and drugs in an individual patient can vary depending on genetic background and the presence of other disorders and drugs that effect lipid/lipoprotein levels. For an extensive discussion of the secondary disorders that alter lipid and lipoprotein metabolism please refer to the individual Endotext chapters on these disorders. For additional information on the effect of drugs on lipid and lipoprotein metabolism please see the Endotext chapter on this topic (5).

 

Table 1. Disorders Associated with an Increase LDL in Cholesterol Levels

Increased intake of saturated or trans fatty acids

Hypothyroidism

Obstructive liver disease

Nephrotic syndrome

Pregnancy

Growth hormone deficiency

Anorexia nervosa

Monoclonal gammopathy

Cushing’s syndrome

Acute intermittent porphyria

Hepatoma

 

Table 2. Drugs That Increase LDL Cholesterol Levels

Cyclosporine and tacrolimus

Amiodarone

Glucocorticoids

Danazol

Some progestins

Protease inhibitors

Anabolic steroids

Androgen deprivation therapy

Retinoids

Thiazide diuretics

Loop diuretics

Thiazolidinediones

SGLT2 inhibitors

 

Table 3. Disorders Associated with an Increase in Triglyceride Levels

Obesity

Alcohol intake

High simple carbohydrate diet

Diabetes

Metabolic syndrome

Polycystic ovary syndrome

Hypothyroidism

Chronic renal failure

Nephrotic syndrome

Pregnancy

Inflammatory diseases (Rheumatoid arthritis, Lupus, psoriasis, etc.)

Infections

Acute stress (myocardial infarctions, burns, etc.)

HIV

Cushing’s syndrome

Growth hormone deficiency

Lipodystrophy

Glycogen Storage disease

Acute hepatitis

Monoclonal gammopathy

 

Table 4. Drugs That Increase Triglyceride Levels

Alcohol

Oral Estrogens

Tamoxifen/Raloxifene

Glucocorticoids

Retinoids

Beta blockers

Thiazide diuretics

Loop diuretics

Protease Inhibitors

Cyclosporine, sirolimus, and tacrolimus

Atypical anti-psychotics

Bile acid sequestrants

L-asparaginase

Androgen deprivation therapy

Cyclophosphamide

Alpha-interferon

Propofol

 

Table 5. Disorders Associated with a Decrease in HDL Cholesterol Levels

Marked Hypertriglyceridemia

Obesity

Metabolic syndrome

Low fat intake

Infection

Inflammation

Malignancy

Severe liver disease

Polycystic ovary syndrome

Paraproteinemia (artifact of some assays)

 

Table 6. Drugs That Decrease HDL Cholesterol Levels

Anabolic steroids

Danazol

TZD + fibrate (idiosyncratic reaction)

Beta-blockers

Progestins

Anti-psychotics

 

Table 7. Disorders or Drugs Associated with an Increase in Lp(a) Levels

Chronic Kidney Disease

Nephrotic Syndrome

Inflammation

Hypothyroidism

Acromegaly

Polycystic ovary syndrome

Growth hormone therapy

Androgen deprivation therapy

Statins

 

In a patient with an elevated LDL-C level, one should take a diet history, review the medication list, and check a TSH level to rule out hypothyroidism. Most of the disorders that cause elevations in LDL-C levels, other than hypothyroidism, should be obvious on routine history, physical examination, and laboratory screening. In a patient with an elevated triglyceride level, one should take a diet history and in particular focus on the ingestion of simple sugars and ethanol. One should review the medication list and recognize that many common disease states can adversely impact triglyceride levels including obesity, poorly controlled diabetes, chronic renal failure, HIV, and inflammatory disorders (4,6-9). Weight loss, improvements in glycemic control in patients with diabetes, and a reduction of inflammation can all result in a decrease in triglyceride levels (4,6,7). In a patient with a low HDL-C level one should review the medication list and diet, recognizing that diets very low in fat can result in low HDL-C levels, which are often accompanied by low LDL-C and triglyceride levels (10). In young or very fit males with very low HDL-C levels a careful history directed at anabolic steroid use is essential (3).

 

THINK ABOUT GENETIC CAUSES

 

One should always consider the possibility of a genetic disorder and therefore ask whether relatives have either premature cardiovascular disease, lipid disorders, or are taking lipid lowering medications (11). If the triglyceride levels are markedly elevated one should inquire about a family history of pancreatitis. When the lipid/lipoprotein abnormality is markedly abnormal or begins at a young age, the likelihood of a genetic disorder is increased and the family history assumes even greater importance. It is essential to think about the possibility of a genetic disorder because many of the common lipid disorders, such as familial hypercholesterolemia and elevations in Lp(a), have an autosomal codominant genetic transmission and therefore will be present in approximately 50% of family members (12-15). The recognition of the possibility of a genetic disorder will lead to screening family members and if abnormalities are found early treatment can be initiated, which may prevent the adverse consequences of hyperlipidemia. The monogenetic disorders that cause elevations in LDL-C and triglycerides levels and low HDL-C levels are shown in tables 8-11.

 

Table 8. Elevation in LDL Cholesterol (Familial Hypercholesterolemia)

LDL receptor mutations

Autosomal codominant

Approx. 1 in 250

Apolipoprotein B mutations

Autosomal codominant

Approx. 1 in 1000

PCSK9 mutations

Autosomal codominant

rare

Autosomal recessive hypercholesterolemia

Autosomal recessive

rare

Lysosomal acid lipase deficiency

Autosomal recessive

rare

Cholesterol 7 alpha hydroxylase deficiency

Autosomal recessive

rare

Sitosterolemia (ABCG5/ABCG8)

Autosomal recessive

rare

In autosomal codominant disorders heterozygotes have lipid abnormalities approximately half as severe as homozygotes

 

Table 9. Marked Elevations in Triglycerides (Familial Chylomicronemia Syndrome)

Lipoprotein lipase deficiency

Autosomal recessive

rare

Apolipoprotein C-II deficiency

Autosomal recessive

rare

Apolipoprotein A-V deficiency

Autosomal recessive

rare

GPIHBP1 deficiency

Autosomal recessive

rare

Lipase maturation factor 1 deficiency

Autosomal recessive

rare

 

Table 10. Elevations in Triglycerides and Cholesterol

Familial Dysbetalipoproteinemia

Apo E2/E2, rare mutations in Apo E

1-5/5000

 

Table 11. Decreased HDL Cholesterol

Apolipoprotein A-I deficiency or variants

Autosomal codominant

rare

Tangier disease (ABCA1 deficiency)

Autosomal codominant

rare

LCAT deficiency

Autosomal codominant

rare

In autosomal codominant disorders heterozygotes have lipid abnormalities approximately half as severe as homozygotes

 

Very frequently hypertriglyceridemia and/or hypercholesterolemia are due to polygenic inheritance secondary to combinations of common small effect genes that regulate the production or catabolism of lipoproteins (16). In addition, lifestyle, other disease states, and medications can interact with genetic susceptibilities to result in marked dyslipidemia and therefore even when a genetic disorder is present one should not ignore reversible factors where appropriate treatment can have marked effects on lipid levels. Often secondary factors facilitate the expression of genetic variations to result in an abnormal lipid phenotype. One of the best examples of the interaction of secondary factors and genetic variants is familial dysbetalipoproteinemia (17,18). The apolipoprotein E2/E2 polymorphism occurs in approximately 1% of individuals whereas the clinical disorder only occurs in 1-5/5000 and is frequently associated with other disorders, such as obesity, hypothyroidism, and diabetes, which also perturb lipid metabolism (17,18). A detailed discussion of the genetic disorders that effect plasma lipid and lipoprotein levels can be found in the individual Endotext chapters that focus on these disorders.

 

ORDERING SPECIAL LABORATORY STUDIES

 

In most circumstances a routine lipid panel consisting of triglycerides, total cholesterol, HDL-C, and calculated LDL-C and non-HDL-C provides sufficient information to appropriately decide on who to treat and the best treatment approach. In a patient with high fasting triglycerides (>200-400mg/dl) where the LDL-C cannot be accurately calculated measurement of direct LDL-C maybe very helpful. However, it should be recognized that there are certain situations where more sophisticated and detailed laboratory studies can be helpful (19). For example, in a young patient with atherosclerotic vascular disease and no obvious risk factors and a normal lipid profile obtaining specialized lipid/lipoprotein studies including measurement of Lp(a) would be indicated. Indications for measuring Lp(a) are shown in Table 12 (19,20). Note, it is the opinion of some experts that Lp(a) should be measured once in all individuals. The various specialized lipid and lipoprotein studies and their appropriate use are discussed in detail in the Endotext chapter “Utility of Advanced Lipoprotein Testing in Clinical Practice” (19). 

 

Table 12. When to Measure Lp(a) Levels

Patients with premature CHD

Patients with a strong family history of premature CHD

Patients with a family history of elevated Lp(a) levels (Cascade screening)

Patients with resistance to LDL-C lowering with statins

Patients with familial hypercholesterolemia

Patients with aortic valvular stenosis of uncertain cause

Patients with an unknown cause of ischemic stroke

Patients with intermediate risk profiles

Note: It is the opinion of some experts that Lp(a) should be measured once in all individuals

 

DECIDING WHO TO TREAT

 

The purpose of treating lipid disorders is to prevent the development of other diseases, particularly cardiovascular disease. Thus, the decision to treat should be based on the risk of the hyperlipidemia leading to those medical problems. A number of guidelines have been published that discuss in detail cardiovascular risk assessment and provide recommendations on treatment strategies (21-24). It should be noted that while these guidelines are similar there are significant differences between their recommendations. Additionally, several calculators are available on-line to determine an individual patient’s risk of developing cardiovascular disease in the next 10 years or their lifetime risk. These issues are discussed in detail in the chapters on Risk Assessment and Guidelines for the Management of High Blood Cholesterol and Triglycerides (25-27). In addition to cardiovascular complications, marked elevations in triglycerides can lead to pancreatitis (26). The National Lipid Association recommends treating triglyceride levels greater than 500mg/dl while the Endocrine Society recommends treating triglycerides if they are greater than 1000mg/dl to lower the risk of pancreatitis (28,29).

 

GOALS OF THERAPY

 

The current American College of Cardiology/American Heart Association (ACC/AHA) guidelines do not emphasize specific lipid/lipoprotein goals of therapy but rather to just treat with the statins to lower LDL-C by a certain percentage (21). An exception is that they do recommend in patients with very high-risk ASCVD, to use an LDL-C threshold of 70 mg/dL to consider addition of non-statins to statin therapy. In contrast, other groups, such as the National Lipid Association, International Atherosclerosis Society, European Society of Cardiology/European Atherosclerosis Society, and AACE, do recommend lowering the LDL and non-HDL cholesterol levels to below certain levels depending upon the cardiovascular risk in a particular patient but the recommendations from these organizations are not identical (22,24,29,30).

 

A detailed discussion of lipid/lipoprotein goals is provided in the chapter on Risk Assessment and Guidelines for the Management of High Blood Cholesterol (25). It should be noted that many lipid experts would recommend trying to achieve an LDL-C levels less than 70mg/dl and non-HDL-C levels less than 100mg/dl in patients with cardiovascular disease or patients at very high risk for the development of cardiovascular disease. In other patients, an LDL-C level less than 100mg/dl and non-HDL-C level less than 130mg/dl is a reasonable goal. AACE and European Society of Cardiology/European Atherosclerosis Society have recommended LDL-C levels less than 55mg/dl in patients at very high risk (22,24). With the results of the IMPROVE-IT trial and PCKS9 inhibitor studies, which showed that adding ezetimibe or a PCSK9 inhibitor to statin therapy resulted in an additional decrease in LDL-C levels and a further reduction in cardiovascular events, the arguments in favor of trying to reach lower lipid/lipoprotein goals has been greatly strengthened  (31-33). Moreover, the results of these and other studies provide strong support that the lower the LDL-C level the greater the reduction in cardiovascular events (34,35).     

 

TREATMENT TO REDUCE COMPLICATIONS OF DYSLIPIDEMIA

 

The first priority in treating lipid disorders is to lower the LDL-C levels to goal, unless triglycerides are markedly elevated (> 500-1000mg/dl), which increases the risk of pancreatitis. LDL-C is the usual first priority because the data linking lowering LDL-C with reducing cardiovascular disease are extremely strong and we now have the ability to markedly decrease LDL-C levels. Dietary therapy is the initial step but in the majority of patients’ dietary modifications will not be sufficient to achieve the LDL-C goals. If patients are willing and able to make major changes in their diet it is possible to achieve remarkable reductions in LDL-C levels but this seldom occurs in clinical practice. Additionally, the dietary changes need to be sustained for a long period of time to be effective and many patients while able to follow an LDL-C lowering diet in the short term are unable to follow the diet for an extended period of time.

 

Primary Prevention Patients

 

The first step is determining the risk for developing atherosclerotic cardiovascular disease. There are a number of different calculators for determining risk. In the US the most popular is the ACC/AHA risk calculator (http://www.cvriskcalculator.com/) whereas in Europe the SCORE (Systematic Coronary Risk Estimation) is popular (SCORE2 and SCORE2-OP (escardio.org)). The ACC/AHA recommendations are shown in Figure 1 and the European Society of Cardiology/European Atherosclerosis Society recommendations are shown in Figure 2.

 

Figure 1. ACC/AHA Recommendations for Patients without ASCVD, Diabetes, or LDL-C greater than 190mg/dl. Risk enhancers are listed in table 13. (Note the risk is for MI and stroke, both fatal and nonfatal).

 

 

Table 13. ASCVD Risk Enhancers

Family history of premature ASCVD
Persistently elevated LDL > 160mg/dl
Chronic kidney disease
Metabolic syndrome
History of preeclampsia
History of premature menopause
Inflammatory disease (especially rheumatoid arthritis, psoriasis, HIV)
Ethnicity (e.g., South Asian ancestry)
Persistently elevated triglycerides > 175mg/dl
Hs-CRP > 2mg/L
Lp(a) > 50mg/dl or >125nmol/L
Apo B > 130mg/dl
Ankle-brachial index (ABI) < 0.9

 

Figure 2. European Society of Cardiology/European Atherosclerosis Society Recommendations for Primary Prevention Patients. Risk categories are shown in table 14. (Note that the SCORE risk is for a fatal event). There are different tables for different European countries.

 

Table 14. Cardiovascular Risk Categories

Very High Risk

ASCVD

DM with target organ damage or at least three major risk factors or early onset of T1DM of long duration (>20 years)

Severe CKD (eGFR <30 mL/min/1.73 m2)

A calculated SCORE >10% for 10-year risk of fatal CVD

FH with ASCVD or with another major risk factor

High Risk

Markedly elevated single risk factors, in particular TC >310 mg/dL, LDL-C >190 mg/dL, or BP >180/110 mmHg

Patients with FH without other major risk factors

Patients with DM without target organ damage with DM duration >10 years or another additional risk factor

Moderate CKD (eGFR 30[1]59 mL/min/1.73 m2).

A calculated SCORE >5% and <10% for 10-year risk of fatal CVD

Moderate Risk

Young patients (T1DM <35 years; T2DM <50 years) with DM duration <10 years, without other risk factors

Calculated SCORE >1 % and <5% for 10-year risk of fatal CVD.

Low Risk

Calculated SCORE <1% for 10-year risk of fatal CVD

 

A few caveats are worth noting. First, in patients less than 60 years of age it is very helpful to calculate the life-time risk of ASCVD events. Often one will find that the 10-year risk is modest but the life-time risk is high and this information should be included in the risk discussion to help in the decision process. Second, patients should be made aware of the natural history of ASCVD i.e., that it begins early in life and slowly progresses overtime with high LDL-C levels accelerating the rate of development of atherosclerosis and low LDL-C leading to a slower progression of atherosclerosis (35,36). Third, patients should be made aware of genetic studies demonstrating that variants in genes that lead to lifetime decreases in LDL-C levels (for example the HMG-CoA reductase gene, NPC1L1 gene, PCSK9 gene, ATP citrate lyase gene, and LDL receptor gene) result in a decreased risk of cardiovascular events. In a recent study it was reported that a 10mg/dL lifetime decrease in LDL-C with any of these genetic variants was associated with a 16-18% decrease in cardiovascular events whereas a 10mg/dl reduction in LDL-C with lipid lowering therapy results in only approximately a 5% decrease in cardiovascular events (34,37,38). The combination of the natural history and the results observed with genetic variants strongly suggests that early therapy to lower LDL-C levels will have greater effects on reducing the risk of ASCVD events than starting therapy later in life. This information needs to be discussed with the patient. Fourth, if the patient or health care provider are uncertain of the best course of action obtaining a cardiac calcium scan can be very helpful in the decision-making process, particularly in older individuals. A score of 0, particularly in an older patient would indicate that statin therapy is not needed whereas a score > 100 would indicate a need for statin therapy (21). A score of 1-99 favors the use of a statin (21).

 

In most primary prevention patients, statin therapy is sufficient to lower LDL-C levels to goal (< 100mg/dl). One can usually start with moderate statin therapy (for example atorvastatin 10-20mg or rosuvastatin 5-10mg) and increase the statin dose, if necessary, to achieve LDL-C goals. Statins are available as generic drugs and therefore are relatively inexpensive. If a patient does not achieve their LDL-C goal on intensive statin therapy, cannot tolerate statin therapy, or is able to take only a low dose of a statin one can use ezetimibe (generic drug), bempedoic acid, bile acid sequestrants, or PCSK9 inhibitors to further lower LDL-C levels (for detailed discussion of cholesterol lowering drugs see (39)). It should be noted that the addition of ezetimibe or a PCSK9 inhibitor to statin therapy has been shown to reduce cardiovascular events (31-33). In most situations, ezetimibe is the drug of choice given its low cost, ability to reduce ASCVD events, and long-term safety record. If LDL-C is not close to goal PCSK9 inhibitors can be used. Once LDL-C is at goal if the non-HDL-C remains high one can consider the approaches described in the section describing the approach to patients with LDL-C at goal with elevated triglycerides.

 

Patients with LDL Cholesterol Greater than 190mg/dl

 

When the LDL-C is greater than 190mg/dl the patient should be started on intensive statin therapy (atorvastatin 40-80mg per day or rosuvastatin 20-40mg per day). If the LDL-C goal is not achieved (usually < 100mg/dl) additional lipid lowering medications should be added. If the LDL-C is relatively close to goal one can use ezetimibe but if the LDL-C is far from the goal the use of a PCSK9 inhibitor should be employed. Because of the potential for a genetic disorder, either monogenic or polygenic, one should check family members for lipid abnormalities. If possible genetic testing for monogenic disorders causing hypercholesterolemia is recommended (40).

 

Patients with Diabetes

 

Most patients with diabetes (age 40-75) without risk factors should be started on moderate statin therapy (for example atorvastatin 10-20mg or rosuvastatin 5-10mg). In young individuals (< age 40) and older individuals (> age 75) one needs to use clinical judgment Patients with diabetes with ASCVD or risk factors should be started on intensive statin therapy. In my opinion reasonable goals are shown in table 15 (similar to AACE and ADA guidelines) (24,41). If intensive statin therapy does not achieve LDL-C goals additional drugs can be added. If reasonably close to the LDL-C goal the initial drug added should be ezetimibe. If far from goal one could add a PCSK9 inhibitor. Once LDL-C is at goal if the non-HDL-C remains high one can consider the approaches described in the section describing the approach to patients with LDL-C at goal with elevated triglycerides.

 

Table 15. ASCVD Risk Categories and Treatment Goals

Risk Category

Risk Factors/10-year risk

LDL-C mg/dl

Non-HDL-C mg/dl

Extreme Risk

Diabetes and clinical cardiovascular disease

<55

<80

Very High Risk

Diabetes with one or more risk factors

<70

<100

High Risk

Diabetes and no other risk factors

<100

<130

 

Secondary Prevention Patients

 

Patients with ASCVD (secondary prevention patients) should be started on intensive statin therapy (atorvastatin 40-80mg per day or rosuvastatin 20-40mg per day). Given the extensive data showing that the lower the LDL-C the greater the reduction in ASCVD events most secondary prevention patients would benefit from the addition of ezetimibe to maximize LDL-C lowering without markedly increasing costs (34,35). The goal LDL-C in this patient population is an LDL<70mg/dl but many experts and some guidelines would prefer an LDL-C<55mg/dl if possible. If on intensive statin therapy and ezetimibe treatment the LDL-C is far above goal one could consider adding a PCSK9 inhibitor (this is particularly necessary if the LDL-C is greater than 100mg/dl or the patient is at very high risk due to other factors (diabetes, cerebral vascular disease, peripheral vascular disease, recent MI, history of multiple MIs) (34,35).

 

Patients with LDL Cholesterol at Goal but High Triglycerides (>150mg/dl to <500mg/dl)

 

Patients with an LDL-C at goal but high triglyceride levels (>150mg/dl to <500mg/dl) will often have increased non-HDL-C levels. Numerous studies have shown that the risk of ASCVD events is increased in this patient population (42). The initial step should be to improve lifestyle, treat secondary disorders that may be contributing to the increase in triglycerides, and if possible, discontinue medications that increase triglyceride levels. Studies have not demonstrated a reduction in cardiovascular events when niacin is added to statin therapy and given the side effects of niacin enthusiasm for using niacin in combination with statins to reduce ASCVD is limited (43,44). Additionally, the ACCORD-LIPID trial failed to demonstrate that adding fenofibrate to statin therapy (45) and the PROMINENT trial failed to demonstrate that adding pemafibrate to statin therapy (46) reduces cardiovascular disease. Thus, there is little evidence that adding either niacin or a fibrate to statin therapy will be beneficial in reducing cardiovascular events.

 

The REDUCE-IT trial demonstrated that adding the omega-3-fatty acid icosapent ethyl (EPA; Vascepa) to statin therapy in patients with elevated triglyceride levels reduced the risk of ASCVD events by 25% while decreasing triglyceride levels by 18% (47). Similar results were seen in the JELIS trial (48). In these trials the reduction in triglyceride levels was relatively modest and would not have been expected to result in the magnitude of the decrease in cardiovascular disease observed in the JELIS and REDUCE-IT trials. Other actions of EPA, such as decreasing platelet function, anti-inflammation, decreasing lipid oxidation, stabilizing membranes, etc. could account for or contribute to the reduction in cardiovascular events (49). Based on these results the National Lipid Association has recommended “that for patients aged ≥45 years with clinical ASCVD, or aged ≥50 years with diabetes mellitus requiring medication plus ≥1 additional risk factor, with fasting TGs 135 to 499 mg/dL on high-intensity or maximally tolerated statin therapy (±ezetimibe), treatment with icosapent ethyl is recommended for ASCVD risk reduction” (50). However, it should be recognized that the STRENGTH trial using a carboxylic acid formulation of EPA and DHA failed to reduce cardiovascular events despite reducing triglyceride levels to a similar degree as in the REDUCE-IT trial (51). Whether EPA has special properties that resulted in the reduction in cardiovascular events in the REDUCE-IT trial or there were flaws in the trial (the use of mineral oil as the placebo) is debated (49,52). For a detailed discussion of triglyceride lowering drugs see the Endotext chapter on this topic (53).

 

Patients with Very High Triglyceride Levels (>500-1000mg/dl)

 

The main aim is to keep triglyceride levels below 500 mg/dL to prevent triglyceride-induced pancreatitis (17,54,55).

 

FAMILIAL HYPERCHYLOMICRONEMIA (FCS)

 

FCS is a rare autosomal recessive disorder due to an abnormality in the genes listed in table 9 that result in the absence of functional lipoprotein lipase (LPL) activity (17,54,55). Patients with FCS respond poorly to most triglyceride lowering drugs (fibrates, omega-3-fatty acids, niacin) (17,54,55). A very low-fat diet (5-10% of total calories) is the most effective treatment but can be difficult for many patients to comply with (17,54,55). Volanesorsen, a drug that is approved in Europe but not in the US, lowers ApoC-III levels and is effective in lowering triglyceride levels in patients with FCS (53).

 

MULTIFACTORIAL CHYLOMICRONEMIA SYNDROME (MCS)

 

MCS is due to the coexistence of a genetic predisposition (polygenic or heterozygous for genes that cause FCS) to hypertriglyceridemia with 1 or more secondary causes of hypertriglyceridemia (see tables 3 and 4) (17,54,55). Initial treatment is a very low-fat diet to reduce triglyceride levels into a safe range (<1000mg/dl). Treating secondary disorders that raise triglyceride levels and when possible, stopping drugs that increase triglyceride levels is essential (17,54,55). If the triglyceride levels remain above 500mg/dl the addition of fenofibrate or omega-3-fatty acids is indicated. Many patients with MCS are at high risk for ASCVD and therefore after triglyceride levels are controlled the patient should be evaluated for cardiovascular disease risk and if indicated statin therapy initiated.   

 

Patients with High Lp(a) Levels

 

Life style changes do not significantly lower Lp(a) levels (56). The effect of lipid lowering drugs on Lp(a) levels is shown in Table 16. In patients with elevations in Lp(a) the initial therapy is to aggressively control the other cardiovascular disease risk factors. In some instances, one can use niacin, PCSK9 inhibitors, or in postmenopausal women estrogen to lower Lp(a) levels but the effect of these drugs on preventing cardiovascular events by lowering Lp(a) levels is uncertain (57). Studies of an antisense oligonucleotide or small interfering RNA (both not yet approved) directed at apo(a) have shown that these drugs can lower Lp(a) by >75% without effecting other lipoprotein levels (58). Lipoprotein apheresis can be employed to lower Lp(a) in patients with very high Lp(a) levels who continue to have cardiovascular events despite optimal medical management (59).  

 

Table 16. Effect of Lipid Lowering Drugs on Lp(a) Levels

Statins

No Effect or slight increase

Ezetimibe

No Effect or slight increase

Fibrates

No Effect

Niacin

Decrease 15-25%. Greatest decrease in patients with highest Lp(a) levels

PCSK9 Inhibitors

Decrease 20-30%

Estrogen

Decrease 20-35%

Mipomersen**

Decrease 25-30%

Lomitapide*

Decrease 15-20%

Evinacumab

No effect in homozygous familiar hypercholesterolemia

Decrease 16% in refractory hypercholesterolemia

CETP Inhibitors**

Decrease ~ 25%

Apo (a) antisense**

Decrease > 75%

*only approved for the treatment of Homozygous FH; **not currently available

 

Decreased HDL Cholesterol Levels

 

Despite epidemiologic studies consistently showing that high HDL-C levels are associated with a decreased risk of cardiovascular disease there are no studies demonstrating that increasing HDL-C levels reduces cardiovascular disease (60). It should be recognized that the crucial issue with HDL may not be the HDL-C levels per se but rather the function of the HDL particles (60). Assays have been developed to determine the ability of HDL to facilitate cholesterol efflux from macrophages and these studies have shown that the levels of HDL-C do not necessarily indicate the ability to mediate cholesterol efflux (61). Similarly, the ability of HDL to protect LDL from oxidation may also play an important role in the ability of HDL to reduce ASCVD (62). Thus, the functional capability of HDL may be more important than HDL-C levels (60-62).

 

CONCLUSION

 

In summary, modern therapy demands that we aggressively evaluate and when indicated treat lipid disorders to reduce the risk of atherosclerotic cardiovascular disease and in those with very high triglycerides to reduce the risk of pancreatitis.

 

REFERENCES

 

  1. Shapiro MD, Feingold KR. Monogenic Disorders Causing Hypobetalipoproteinemia. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Hofland J, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2021.
  2. Shapiro MD, Feingold KR. Monogenic Disorders Altering HDL Levels. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Hofland J, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2021.
  3. Feingold KR, Brinton EA, Grunfeld C. The Effect of Endocrine Disorders on Lipids and Lipoproteins. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Hofland J, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2020.
  4. Feingold KR. Diabetes and Dyslipidemia. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Hofland J, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2020.
  5. Herink M, Ito MK. Medication Induced Changes in Lipid and Lipoproteins. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Hofland J, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2018.
  6. Feingold KR. Obesity and Dyslipidemia. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Hofland J, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2020.
  7. Feingold KR, Grunfeld C. The Effect of Inflammation and Infection on Lipids and Lipoproteins. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Hofland J, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2022.
  8. K. R, Tannock L. Dyslipidemia in Chronic Kidney Disease. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Hofland J, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2022.
  9. Sarkar S, Brown TT. Lipid Disorders in People with HIV. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Hofland J, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2023.
  10. Feingold KR. The Effect of Diet on Cardiovascular Disease and Lipid and Lipoprotein Levels. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Hofland J, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2021.
  11. Patni N, Ahmad Z, Wilson DP. Genetics and Dyslipidemia. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Hofland J, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2020.
  12. Levenson AE, de Ferranti SD. Familial Hypercholesterolemia. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Hofland J, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2020.
  13. McNeal CJ, Peterson AL. Lipoprotein (a) in Youth. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Hofland J, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2020.
  14. Warden BA, Fazio S, Shapiro MD. Familial Hypercholesterolemia: Genes and Beyond. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Hofland J, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2021.
  15. Hegele RA, Boren J, Ginsberg HN, Arca M, Averna M, Binder CJ, Calabresi L, Chapman MJ, Cuchel M, von Eckardstein A, Frikke-Schmidt R, Gaudet D, Hovingh GK, Kronenberg F, Lutjohann D, Parhofer KG, Raal FJ, Ray KK, Remaley AT, Stock JK, Stroes ES, Tokgozoglu L, Catapano AL. Rare dyslipidaemias, from phenotype to genotype to management: a European Atherosclerosis Society task force consensus statement. Lancet Diabetes Endocrinol 2020; 8:50-67
  16. Dron JS, Hegele RA. Polygenic influences on dyslipidemias. Curr Opin Lipidol 2018; 29:133-143
  17. Chait A, Subramanian S. Hypertriglyceridemia: Pathophysiology, Role of Genetics, Consequences, and Treatment. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Hofland J, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2019.
  18. Shah AS, Wilson DP. Genetic Disorders Causing Hypertriglyceridemia in Children and Adolescents. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Hofland J, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2020.
  19. Feingold KR. Utility of Advanced Lipoprotein Testing in Clinical Practice. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Hofland J, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2023.
  20. Wilson DP, Jacobson TA, Jones PH, Koschinsky ML, McNeal CJ, Nordestgaard BG, Orringer CE. Use of Lipoprotein(a) in clinical practice: A biomarker whose time has come. A scientific statement from the National Lipid Association. J Clin Lipidol 2022; 16:e77-e95
  21. Grundy SM, Stone NJ, Bailey AL, Beam C, Birtcher KK, Blumenthal RS, Braun LT, de Ferranti S, Faiella-Tommasino J, Forman DE, Goldberg R, Heidenreich PA, Hlatky MA, Jones DW, Lloyd-Jones D, Lopez-Pajares N, Ndumele CE, Orringer CE, Peralta CA, Saseen JJ, Smith SC, Jr., Sperling L, Virani SS, Yeboah J. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA Guideline on the Management of Blood Cholesterol: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 2019; 139:e1082-e1143
  22. Mach F, Baigent C, Catapano AL, Koskinas KC, Casula M, Badimon L, Chapman MJ, De Backer GG, Delgado V, Ference BA, Graham IM, Halliday A, Landmesser U, Mihaylova B, Pedersen TR, Riccardi G, Richter DJ, Sabatine MS, Taskinen MR, Tokgozoglu L, Wiklund O, E. S. C. Scientific Document Group. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: lipid modification to reduce cardiovascular risk. Eur Heart J2020; 41:111-188
  23. U. S. Preventive Services Task Force, Mangione CM, Barry MJ, Nicholson WK, Cabana M, Chelmow D, Coker TR, Davis EM, Donahue KE, Jaen CR, Kubik M, Li L, Ogedegbe G, Pbert L, Ruiz JM, Stevermer J, Wong JB. Statin Use for the Primary Prevention of Cardiovascular Disease in Adults: US Preventive Services Task Force Recommendation Statement. JAMA 2022; 328:746-753
  24. Handelsman Y, Jellinger PS, Guerin CK, Bloomgarden ZT, Brinton EA, Budoff MJ, Davidson MH, Einhorn D, Fazio S, Fonseca VA, Garber AJ, Grunberger G, Krauss RM, Mechanick JI, Rosenblit PD, Smith DA, Wyne KL. Consensus Statement by the American Association of Clinical Endocrinologists and American College of Endocrinology on the Management of Dyslipidemia and Prevention of Cardiovascular Disease Algorithm - 2020 Executive Summary. Endocr Pract 2020; 26:1196-1224
  25. Grundy SM, Feingold KR. Guidelines for the Management of High Blood Cholesterol. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Hofland J, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2022.
  26. Tannock L, RR G. Risk of Fasting and Non-Fasting Hypertriglyceridemia in Coronary Vascular Disease and Pancreatitis. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Hofland J, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson D, eds. Endotext. South Dartmouth (MA) 2022.
  27. Daniels SR. Guidelines for Screening, Prevention, Diagnosis and Treatment of Dyslipidemia in Children and Adolescents. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Hofland J, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2023.
  28. Berglund L, Brunzell JD, Goldberg AC, Goldberg IJ, Sacks F, Murad MH, Stalenhoef AF, Endocrine Society. Evaluation and treatment of hypertriglyceridemia: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 2012; 97:2969-2989
  29. Jacobson TA, Ito MK, Maki KC, Orringer CE, Bays HE, Jones PH, McKenney JM, Grundy SM, Gill EA, Wild RA, Wilson DP, Brown WV. National Lipid Association recommendations for patient-centered management of dyslipidemia: part 1 - executive summary. J Clin Lipidol 2014; 8:473-488
  30. Expert Panel on Dyslipidemia. An International Atherosclerosis Society position paper: global recommendations for the management of dyslipidemia: executive summary. Atherosclerosis 2014; 232:410-413
  31. Cannon CP, Blazing MA, Giugliano RP, McCagg A, White JA, Theroux P, Darius H, Lewis BS, Ophuis TO, Jukema JW, De Ferrari GM, Ruzyllo W, De Lucca P, Im K, Bohula EA, Reist C, Wiviott SD, Tershakovec AM, Musliner TA, Braunwald E, Califf RM, Improve-It Investigators. Ezetimibe Added to Statin Therapy after Acute Coronary Syndromes. N Engl J Med 2015; 372:2387-2397
  32. Sabatine MS, Giugliano RP, Keech AC, Honarpour N, Wiviott SD, Murphy SA, Kuder JF, Wang H, Liu T, Wasserman SM, Sever PS, Pedersen TR, Fourier Steering Committee

         Investigators,. Evolocumab and Clinical Outcomes in Patients with Cardiovascular Disease. N Engl J Med 2017; 376:1713-1722

  1. Schwartz GG, Steg PG, Szarek M, Bhatt DL, Bittner VA, Diaz R, Edelberg JM, Goodman SG, Hanotin C, Harrington RA, Jukema JW, Lecorps G, Mahaffey KW, Moryusef A, Pordy R, Quintero K, Roe MT, Sasiela WJ, Tamby JF, Tricoci P, White HD, Zeiher AM, Odyssey Outcomes Committees Investigators. Alirocumab and Cardiovascular Outcomes after Acute Coronary Syndrome. N Engl J Med 2018; 379:2097-2107
  2. Feingold KR. Maximizing the benefits of cholesterol-lowering drugs. Curr Opin Lipidol 2019; 30:388-394
  3. Feingold KR, Chait A. Approach to patients with elevated low-density lipoprotein cholesterol levels. Best Pract Res Clin Endocrinol Metab 2022:101658. PMID: 35487874
  4. Wilson DP. Is Atherosclerosis a Pediatric Disease? In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Hofland J, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2023.
  5. Ference BA, Ray KK, Catapano AL, Ference TB, Burgess S, Neff DR, Oliver-Williams C, Wood AM, Butterworth AS, Di Angelantonio E, Danesh J, Kastelein JJP, Nicholls SJ. Mendelian Randomization Study of ACLY and Cardiovascular Disease. N Engl J Med 2019; 380:1033-1042
  6. Cholesterol Treatment Trialists Collaboration, Baigent C, Blackwell L, Emberson J, Holland LE, Reith C, Bhala N, Peto R, Barnes EH, Keech A, Simes J, Collins R. Efficacy and safety of more intensive lowering of LDL cholesterol: a meta-analysis of data from 170,000 participants in 26 randomised trials. Lancet 2010; 376:1670-1681
  7. Feingold KR. Cholesterol Lowering Drugs. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Hofland J, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2021.
  8. Berberich AJ, Hegele RA. Genetic testing in dyslipidaemia: An approach based on clinical experience. Best Pract Res Clin Endocrinol Metab 2022:101720. PMID: 36682941
  9. ElSayed NA, Aleppo G, Aroda VR, Bannuru RR, Brown FM, Bruemmer D, Collins BS, Das SR, Hilliard ME, Isaacs D, Johnson EL, Kahan S, Khunti K, Kosiborod M, Leon J, Lyons SK, Perry ML, Prahalad P, Pratley RE, Seley JJ, Stanton RC, Gabbay RA, on behalf of the American Diabetes Association. 10. Cardiovascular Disease and Risk Management: Standards of Care in Diabetes-2023. Diabetes Care 2023; 46:S158-S190
  10. Sandesara PB, Virani SS, Fazio S, Shapiro MD. The Forgotten Lipids: Triglycerides, Remnant Cholesterol, and Atherosclerotic Cardiovascular Disease Risk. Endocr Rev 2019; 40:537-557
  11. Hps Thrive Collaborative Group, Landray MJ, Haynes R, Hopewell JC, Parish S, Aung T, Tomson J, Wallendszus K, Craig M, Jiang L, Collins R, Armitage J. Effects of extended-release niacin with laropiprant in high-risk patients. N Engl J Med 2014; 371:203-212
  12. Aim-High Investigators, Boden WE, Probstfield JL, Anderson T, Chaitman BR, Desvignes-Nickens P, Koprowicz K, McBride R, Teo K, Weintraub W. Niacin in patients with low HDL cholesterol levels receiving intensive statin therapy. N Engl J Med 2011; 365:2255-2267
  13. ACCORD Study Group, Ginsberg HN, Elam MB, Lovato LC, Crouse JR, 3rd, Leiter LA, Linz P, Friedewald WT, Buse JB, Gerstein HC, Probstfield J, Grimm RH, Ismail-Beigi F, Bigger JT, Goff DC, Jr., Cushman WC, Simons-Morton DG, Byington RP. Effects of combination lipid therapy in type 2 diabetes mellitus. N Engl J Med 2010; 362:1563-1574
  14. Das Pradhan A, Glynn RJ, Fruchart JC, MacFadyen JG, Zaharris ES, Everett BM, Campbell SE, Oshima R, Amarenco P, Blom DJ, Brinton EA, Eckel RH, Elam MB, Felicio JS, Ginsberg HN, Goudev A, Ishibashi S, Joseph J, Kodama T, Koenig W, Leiter LA, Lorenzatti AJ, Mankovsky B, Marx N, Nordestgaard BG, Pall D, Ray KK, Santos RD, Soran H, Susekov A, Tendera M, Yokote K, Paynter NP, Buring JE, Libby P, Ridker PM, Prominent Investigators. Triglyceride Lowering with Pemafibrate to Reduce Cardiovascular Risk. N Engl J Med2022; 387:1923-1934
  15. Bhatt DL, Steg PG, Miller M, Brinton EA, Jacobson TA, Ketchum SB, Doyle RT, Jr., Juliano RA, Jiao L, Granowitz C, Tardif JC, Ballantyne CM, REDUCE-IT Investigators. Cardiovascular Risk Reduction with Icosapent Ethyl for Hypertriglyceridemia. N Engl J Med 2019; 380:11-22
  16. Yokoyama M, Origasa H, Matsuzaki M, Matsuzawa Y, Saito Y, Ishikawa Y, Oikawa S, Sasaki J, Hishida H, Itakura H, Kita T, Kitabatake A, Nakaya N, Sakata T, Shimada K, Shirato K, Japan, E. P. A. lipid intervention study Investigators. Effects of eicosapentaenoic acid on major coronary events in hypercholesterolaemic patients (JELIS): a randomised open-label, blinded endpoint analysis. Lancet 2007; 369:1090-1098
  17. Mason RP, Sherratt SCR, Eckel RH. Omega-3-fatty acids: Do they prevent cardiovascular disease? Best Pract Res Clin Endocrinol Metab 2022:101681. PMID: 35739003
  18. Orringer CE, Jacobson TA, Maki KC. National Lipid Association Scientific Statement on the use of icosapent ethyl in statin-treated patients with elevated triglycerides and high or very-high ASCVD risk. J Clin Lipidol 2019; 13:860-872
  19. Nicholls SJ, Lincoff AM, Garcia M, Bash D, Ballantyne CM, Barter PJ, Davidson MH, Kastelein JJP, Koenig W, McGuire DK, Mozaffarian D, Ridker PM, Ray KK, Katona BG, Himmelmann A, Loss LE, Rensfeldt M, Lundstrom T, Agrawal R, Menon V, Wolski K, Nissen SE. Effect of High-Dose Omega-3 Fatty Acids vs Corn Oil on Major Adverse Cardiovascular Events in Patients at High Cardiovascular Risk: The STRENGTH Randomized Clinical Trial. JAMA 2020; 324:2268-2280
  20. Goff ZD, Nissen SE. N-3 polyunsaturated fatty acids for cardiovascular risk. Curr Opin Cardiol 2022; 37:356-363
  21. Feingold KR. Triglyceride Lowering Drugs. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Hofland J, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2021.
  22. Chait A, Eckel RH. The Chylomicronemia Syndrome Is Most Often Multifactorial: A Narrative Review of Causes and Treatment. Ann Intern Med 2019; 170:626-634
  23. Chait A, Feingold KR. Approach to patients with hypertriglyceridemia. Best Pract Res Clin Endocrinol Metab2022:101659. PMID: 35459627
  24. Enkhmaa B, Anuurad E, Berglund L. Lipoprotein (a): impact by ethnicity and environmental and medical conditions. J Lipid Res 2016; 57:1111-1125
  25. van Capelleveen JC, van der Valk FM, Stroes ES. Current therapies for lowering lipoprotein (a). J Lipid Res2016; 57:1612-1618
  26. Wei T, Cho L. Recent lipoprotein(a) trials. Curr Opin Lipidol 2022; 33:301-308
  27. Feingold KR. Lipoprotein Apheresis. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Hofland J, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2023.
  28. Thomas SR, Zhang Y, Rye KA. The pleiotropic effects of high-density lipoproteins and apolipoprotein A-I. Best Pract Res Clin Endocrinol Metab 2022:101689. PMID: 36008277
  29. Hovingh GK, Rader DJ, Hegele RA. HDL re-examined. Curr Opin Lipidol 2015; 26:127-132
  30. Navab M, Reddy ST, Van Lenten BJ, Anantharamaiah GM, Fogelman AM. The role of dysfunctional HDL in atherosclerosis. J Lipid Res 2009; 50 Suppl:S145-149

 

Lipoprotein Apheresis

ABSTRACT

 

Lipoprotein apheresis involves the physical removal of lipoproteins from the blood and is employed only in patients where lifestyle and pharmacologic treatment is not capable of decreasing lipoproteins to acceptable levels. There are a number of different guidelines for the use of lipoprotein apheresis. In general, apheresis is indicated for patients with homozygous or heterozygous Familial Hypercholesterolemia (FH) and LDL cholesterol levels >300mg/dl, patients with heterozygous FH and high cardiovascular disease risk characteristics with an LDL cholesterol > 200mg/dl, patients with heterozygous FH and cardiovascular disease or diabetes with an LDL cholesterol > 160mg/dl, or patients with progressive cardiovascular disease and Lp(a) concentrations > 60 mg/dl. Lipoproteins may be removed from the circulation by precipitation, adsorption, or filtration. A number of different systems are currently available for lipoprotein apheresis (heparin precipitation, binding to polyacrylate anions or dextran sulfate, filters that remove lipoproteins based on size, and columns containing antibodies to apolipoprotein B or lipoprotein (a)). The effect of these different lipoprotein apheresis methods on LDL cholesterol and lipoprotein (a) (Lp(a)) levels are very similar with LDL cholesterol and LP(a) levels decreasing by 50-75%. Over 8-13 days the LDL cholesterol and Lp(a) levels increase and may return to baseline levels but in some patients the baseline levels are reduced below the starting values. Triglyceride levels decrease by approximately 50% and HDL cholesterol levels may also decrease depending on the method of apheresis. Triglyceride and HDL cholesterol levels return to baseline levels by 24 hours. Lipoprotein apheresis is generally well tolerated. There are no large randomized studies examining the effect of apheresis on cardiovascular events but there are other types of studies demonstrating the benefits of lipoprotein apheresis on atherosclerosis and cardiovascular disease including observational studies in patients with homozygous FH, studies examining the effect of apheresis on the progression of atherosclerosis, and studies comparing rates of cardiovascular events pre-apheresis and during apheresis. While these studies do not definitively demonstrate that lipoprotein apheresis decreases cardiovascular events, the results of these studies coupled with the randomized studies of LDL cholesterol lowering with drugs makes it extremely likely that lowering LDL cholesterol levels with lipoprotein apheresis will reduce the risk of cardiovascular events. Whether lowering Lp(a) levels with lipoprotein apheresis will reduce cardiovascular events is less certain but given the epidemiology data, genetic studies, basic science experiments, and animal experiments it is very likely that lowering elevated Lp(a) will also have beneficial effects on cardiovascular outcomes. Thus, in carefully selected patient’s lipoprotein apheresis is a useful procedure to lower LDL cholesterol and Lp(a) levels thereby reducing the risk of cardiovascular events. Finally, plasmapheresis has been utilized to rapidly decrease plasma triglyceride levels in patients with very high triglyceride levels and pancreatitis.

 

INTRODUCTION

 

Lipoprotein apheresis involves the physical removal of lipoproteins from the blood and is employed in patients where lifestyle and pharmacologic treatment is not capable of decreasing lipoproteins to acceptable levels (1-4). Lipoprotein apheresis is not widely used but in selected patients can have dramatic effects on lipoprotein levels and clinical benefit (1-4).

 

INDICATIONS

 

Lipoprotein apheresis is only recommended after maximal lifestyle and drug treatment fails to achieve acceptable lipoprotein levels. There are a number of guidelines and recommendations for the use of lipoprotein apheresis. In the United States the Food and Drug Administration has approved the use of lipoprotein apheresis for a limited number of patient categories (Table 1).

 

Table 1. Patients Approved for Lipoprotein Apheresis by the FDA (Kaneka Medical Products Package Information for Liposorber LA 15 system)

1)    1) Familial Hypercholesterolemia homozygotes with LDLc > 500mg/dl

2)    2) Familial Hypercholesterolemia heterozygotes with LDL > 300mg/dl

3)    3) Familial Hypercholesterolemia heterozygotes with LDL > 160mg/dl with coronary heart disease

Patients must be on diet and maximally tolerated drug therapy for 6 months

 

In other countries the guidelines are more liberal. For example, in Germany lipoprotein apheresis is accepted for additional indications (Table 2) (5).

 

Table 2. Indications for Lipoprotein Apheresis in Germany

1)    1) Primary Prevention: patients suffering from FH with LDL cholesterol > 160 mg/dl and cardiovascular events in close relatives.

2)    2) Secondary Prevention: patients with progressive cardiovascular events and LDL cholesterol concentrations > 120–130 mg/dl.

3)    3) Lp(a): independent of LDL cholesterol concentrations patients with progressive cardiovascular disease and Lp(a) concentrations > 60 mg/dl.

Initiation of a lipid apheresis treatment should be considered when diet and lipid lowering

drugs are ineffective

 

In Japan lipoprotein apheresis is approved for patients with coronary artery disease and a total cholesterol level > 250mg/dl (6). The National Lipid Association Recommendations are shown in Table 3 (7).

 

Table 3. National Lipid Association Recommendations for Lipoprotein Apheresis

LDL apheresis may be considered for the following patients who, after 6 months, do not have an adequate response to maximum tolerated drug therapy:

1)    1) Functional homozygous FH with LDL-C ≥300 mg/dL (or non-HDL-C ≥330 mg/dL)

2)    2) Functional heterozygous FH with LDL-C ≥300 mg/dL (or non-HDL-C ≥330 mg/dL) and 0 to 1 risk factors

3)    3) Functional heterozygous FH with LDL-C ≥200 mg/dL (or non-HDL-C ≥230 mg/dL) and high- risk characteristics, such as 2 risk factors or high Lp(a) ≥50 mg/dL using an isoform insensitive assay

4)    4) Functional heterozygous FH with LDL-C ≥160 mg/dL (or non-HDL-C ≥190 mg/dL) and very high-risk characteristics (established CHD, other cardiovascular disease, or diabetes)

 

In general, patients with homozygous Familial Hypercholesterolemia who do not have an adequate response to lipid lowering drugs are candidates for lipoprotein apheresis and this should be initiated as soon as possible. Additionally, apheresis can be considered in patients with elevated cholesterol levels if atherosclerotic vascular disease is present and progressive and if LDL cholesterol treatment goals are not achieved despite maximal drug therapy. The use of lipoprotein apheresis solely for the lowering of Lp(a) is uncertain.

 

In the United States the widespread use of lipoprotein apheresis is limited by the high expense of this treatment and by the small number of centers that perform this procedure (in the US fewer than 60 centers with approximately 600 patients) (2). In contrast, in Germany there are over 350 centers that perform lipoprotein apheresis and the number of patients treated is over 3,000 (4,8).

 

In pregnant women with homozygous or heterozygous Familial Hypercholesterolemia lipoprotein apheresis when available can be utilized to lower LDL cholesterol levels as the use of many drugs is relatively contraindicated during pregnancy (9). In children with homozygous familiar hypercholesterolemia and very high LDL cholesterol levels lipoprotein apheresis treatment can be initiated prior to puberty (10,11).    

 

It is likely that in the future the need for lipoprotein apheresis will be markedly diminished by the recent development of new drugs for lowering LDL cholesterol levels (12). For example, in patients with heterozygous FH the use of PCSK9 inhibitors will markedly reduce the need for lipoprotein apheresis (12). In patients with heterozygous Familial Hypercholesterolemia on lipoprotein apheresis treatment with a PCSK9 inhibitor resulted in 63% to 77% being able to discontinue lipoprotein apheresis (12,13). In patients with homozygous FH the availability of PCSK9 inhibitors, lomitapide, and  evinacumab might also decrease the need for lipoprotein apheresis (12). Additionally, in the future drugs that specifically and markedly lower Lp(a) may become available (14,15). Thus, the number of patients that require lipoprotein apheresis should be limited with the majority of patients having homozygous FH.

 

LIPOPROTEIN APHERESIS METHODS

 

Lipoproteins may be removed from the circulation by precipitation, adsorption, or filtration (Table 4) (2-4,8). A number of different systems are currently available for lipoprotein apheresis (Table 4) (2-4,8).

 

Table 4. Lipoprotein Apheresis Systems

HELP: Heparin-induced extracorporal LDL precipitation

Based on the precipitation of apolipoprotein B containing lipoproteins in acidic conditions by forming complexes with other proteins

DALI: Direct adsorption of lipoproteins

Positively charged apolipoprotein B binds to negatively charged polyacrylate anions

Liposorber: Dextran sulfate

Positively charged apolipoprotein B binds to negatively charged dextran sulfate

MONET: Lipid filtration

Series of filters eliminate lipoproteins based on size

TheraSorb: Apolipoprotein B antibodies

Plasma is passed through columns containing apolipoprotein B antibodies that bind lipoproteins

Lipopac: Apoprotein (a) antibodies

(this is only used for research purposes)

Plasma is passed through columns containing apoprotein (a) antibodies that bind Lp(a)

 

Lipoprotein apheresis is typically carried out on a weekly or biweekly schedule. A typical session is 1.5 – 4 hours. Venous blood is utilized and anticoagulation is required. Some methods utilize plasma (immunoadsorption, filtration, dextran sulfate (Liposorber), HELP) while others utilize whole blood (DALI and dextran sulfate (Liposorber D)) (2-4,8). In the United States HELP precipitation and dextran sulfate adsorption (Liposorber) are approved by the FDA (2). A schematic of the Liposorber system is shown in Figure 1.

 

Figure 1. Liposorber System (http://www.accessdata.fda.gov/cdrh_docs/pdf12/H120005b.pdf)

EFFECT OF LIPOPROTEIN APHERESIS ON LIPOPROTEINS

 

While there are several different lipoprotein apheresis methods (see table 4), the effect of these different methods on plasma lipoprotein levels are similar except for modest differences in their effect on HDL cholesterol levels (2-4,16-18). Soon after lipoprotein apheresis, LDL cholesterol and lipoprotein (a) (Lp(a)) levels are decreased by 50-75% (2-4,16). Over 8-13 days the LDL cholesterol and Lp(a) levels increase such that they may be only modestly decreased or return to baseline prior to the next lipoprotein apheresis session (2,4,19,20). Lp(a) levels tend to rebound more slowly than LDL (2,4,20). After chronic lipoprotein apheresis the pretreatment levels of LDL and Lp(a) in some patients may be reduced by 20 to 40% (2,4,20). Weekly apheresis is more effective in lowering baseline lipoprotein levels than biweekly apheresis. The concomitant use of drug therapy is beneficial, slowing the rebound in lipoprotein levels, even in patients with homozygous FH (2,4,20). In a systemic review of children with homozygous familiar hypercholesterolemia lipoprotein apheresis resulted in a 60-70% reduction in LDL cholesterol (21).

 

Triglyceride levels may decrease by 50% with lipoprotein apheresis but the plasma triglyceride levels return to baseline levels by 24 hours (2,4). HDL cholesterol levels also may transiently decrease by 5-20% but return to baseline within 24 hours (2,4). The explanation for the decrease in HDL cholesterol is uncertain, but may be due to hemodilution, activation of hepatic triglyceride lipase, or the decreased activity of LCAT (2). Notably the acute decrease in HDL cholesterol is greater than the decrease in apolipoprotein A-I (2).

 

Table 5. Effect of Lipoprotein Apheresis on Plasma Lipid and Lipoprotein Levels

Total cholesterol

     ↓↓↓

LDL cholesterol

     ↓↓↓

Lp(a)

     ↓↓↓

HDL cholesterol

     ↓

Triglycerides

     ↓↓

 

The commonly used lipoprotein apheresis methods are not typically used to remove chylomicrons. Instead in patients with markedly elevated triglycerides and severe pancreatitis plasma exchange may be used to rapidly remove chylomicrons and lower plasma triglyceride levels (22,23). 

 

TARGET LEVELS OF LIPOPROTEINS DURING LONG TERM LIPOPROTEIN APHERESIS

 

The following goals of therapy have been suggested (Table 6) (20). It should be recognized that these goals are not based on randomized controlled outcome trials but are suggestions by experts.

 

Table 6. Lipoprotein Targets During Long Term Apheresis

Patient Group

Lipoprotein

Baseline+ (% decrease*)

Interval Mean+ (% decrease*)

FH Homozygote

LDLc

<332mg/dl (>55)

<254mg/dl (>65)

FH Heterozygote

LDLc

---

<101mg/dl (>60)

Increased Lp(a)

Lp(a)

---

<50mg/dl

*Compared with baseline off all lipid lowering treatment

+Baseline levels are immediately before apheresis and interval mean is the level obtained by integrating the area under the post apheresis rebound curve.

 

PLEOTROPIC EFFECTS OF LIPOPROTEIN APHERESIS

 

In addition to decreasing lipoprotein levels, lipoprotein apheresis has other effects (Table 7) (1,2,24).

 

Table 7. Pleotropic Effects of Lipoprotein Apheresis

Decrease in C-reactive protein, SAA, and other inflammatory markers

Decrease in fibrinogen and other coagulation factors

Decrease in plasminogen and other fibrinolytic proteins

Decrease complement

Decrease in plasma and blood viscosity

Decrease in PCSK9 levels

 

It should be noted that the levels of these proteins rapidly return towards normal and the clinical significance of these changes is unknown.

 

EFFECT OF LIPOPROTEIN APHERESIS ON ATHEROSCLEROSIS AND CARDIOVASCULAR OUTCOMES

 

There are no large randomized outcome studies examining the effect of lipoprotein apheresis on cardiovascular event rates. Performing such a study would be very difficult and given the abundance of evidence that marked hypercholesterolemia causes cardiovascular events randomizing patients with very high levels of LDL cholesterol to a group that is not treated would raise ethical concerns. However, there are a large number of other types of studies that provide insights into the benefits of lipoprotein apheresis on atherosclerosis and cardiovascular events.

 

Observational Studies in Patients with Homozygous FH

 

In 1985 Thompson and colleagues reported that plasma exchange for a mean of 8.4 years decreased peak serum cholesterol levels by 37% in five patients with homozygous FH and resulted in 5.5 year longer survival than their five respective homozygous siblings (25). In a larger group of patients with homozygous FH, Keller also reported that survival was improved in the patients treated with lipoprotein apheresis compared to those treated only with drug therapy (26). Additionally, angiographic studies demonstrated that plasma exchange delays the rate of progression of coronary atherosclerosis in homozygotes FH patients (27).

 

Studies Examining the Effect of Lipoprotein Apheresis on Atherosclerosis

 

Several studies have examined the effect of apheresis on atherosclerosis. In 1992 Tatami and colleagues reported that lipoprotein apheresis for greater than one year in 37 patients with hypercholesterolemia (7 homozygote and 25 heterozygote FH patients and 5 undefined patients) had favorable effects on coronary artery stenosis (28). As expected, lipoprotein apheresis decreased LDL cholesterol levels. Definite regression was observed in 14 patients, including 4 homozygotes and 10 heterozygotes and regression was observed in patients with severe or mild atherosclerosis. Moreover, the greater the difference in pre and post LDL cholesterol levels the greater the regression in atherosclerosis. Interestingly patients with other risk factors in addition to hypercholesterolemia had less regression.

 

In 1994 Schuff-Werner and colleagues prospectively determined the efficacy of lipoprotein apheresis in 39 patients with elevated LDL cholesterol levels (286mg/dl) not on statin therapy over a 2 year period (29). Lipoprotein apheresis resulted in a rapid decrease in LDL cholesterol levels from 286mg/dl to 121mg/dl one day after apheresis. Moreover, after one and two years of lipoprotein apheresis the baseline LDL cholesterol levels decreased to 203mg/dl and 205mg/dl, respectively. Angiographic studies were obtained in 33 patients before and after 2 years and demonstrated that the mean degree of stenosis of all segments decreased from 32.5% to 30.6% over the 2 years of apheresis treatment (p=0.02). Additionally, regression > 8% was observed in 50/187 (26.7%) segments, 29/187 (15.5%) segments showed progression, and 108/187 (57.8%) segments were stable (< 8% deviation) over 2 years. Finally, the percentage of patients with angina decreased with lipoprotein apheresis.

 

Waidner and colleagues determined the effect of 3 years of lipoprotein apheresis on coronary artery disease in 32 patients with drug refractory FH (30). Apheresis did not significantly improve exercise tolerance. However, quantitative measurement of 111 circumscribed coronary stenoses showed a mean stenosis of 45 +/- 26% at baseline and 43 +/- 22% after apheresis demonstrating no significant improvement with lipoprotein apheresis.

 

In 1998 Richter and colleagues described the effect of lipoprotein apheresis in 34 patients with coronary heart disease and heterozygous FH not adequately responsive to lipid-lowering drugs (31). Baseline LDL cholesterol levels were 269 +/- 62 mg/dl and the calculated-on treatment interval mean LDL cholesterol was 129 +/- 23mg/dl. Coronary angiography revealed regression of lesions in 4 patients (11.8%) and no progression in 19 patients (55.8%).

 

In a multicenter study Stefanutti et al reported on the effect of lipoprotein apheresis on the progression of coronary artery lesions in 19 patients (32). The levels of LDL cholesterol decreased from 130mg/dl pre-apheresis to 41mg/dl post apheresis. Similarly, Lp(a) levels pre-apheresis decreased from 125mg/dl to 34mg/dl post apheresis. Of note, during apheresis both the pre-apheresis LDL cholesterol and Lp(a) levels were lower than baseline values (LDL: 152mg/dl decreasing to 130mg/dl; Lp(a) 172mg/dl decreasing to 125mg/dl). Coronary catheterization revealed that 94.5% of the lesions were stable over 3.1 years.

 

In 2022 Safarova et al reported the results of lipoprotein apheresis for 10 plus years on carotid intima medial thickness (CIMT) in 10 patients with severe hypercholesterolemia (33). Pretreatment LDL cholesterol was 214mg/dL and 40% of the patients had an Lp(a) >60 mg/dL. As expected, LDL cholesterol and Lp(a) levels decreased (over 70% decrease immediately after apheresis). The percentage of patients with CIMT above their "vascular age" decreased from 80% to 30% over the treatment course and the estimated annual rate of change in mean common CIMT was  minus 4 µm/year.

 

In general, these angiographic studies suggest that lipoprotein apheresis has beneficial effects on coronary artery atherosclerosis. It should be recognized that in many of the patients in the studies described above one would expect worsening of coronary atherosclerosis and therefore the lack of progression in these patients suggests benefit. That these studies demonstrate either regression or decreased progression in these high-risk patients indicates lipoprotein apheresis is having beneficial effects on atherosclerosis.

 

Studies Comparing Pre-Lipoprotein Apheresis Cardiovascular Event Rates to Cardiovascular Event Rates During Lipoprotein Apheresis

 

A number of small studies have compared the rate of cardiovascular events prior to the initiation of lipoprotein apheresis with the rate of cardiovascular events during lipoprotein apheresis treatment. These studies have consistently shown that the rate of cardiovascular events is reduced during apheresis. A larger German Registry study also found evidence supporting a reduction in cardiovascular events during apheresis.

 

STUDIES FOCUSING ON LDL CHOLESTEROL

 

Gordon and colleagues reported the long term effects of lipoprotein apheresis in 49 patients with homozygous (n=10) or heterozygous FH (n=39) (34). As expected, there was a 76% decrease in LDL cholesterol levels immediately following apheresis and in patients with homozygous FH there was a progressive decrease in pretreatment LDL cholesterol levels. In patients with heterozygous FH there was no change in pretreatment LDL cholesterol levels. The rate of cardiovascular events during therapy with LDL apheresis and lipid-lowering drugs was 3.5 events per 1,000 patient-months of treatment compared with 6.3 events per 1,000 patient-months for the 5 years before LDL apheresis therapy (P=0.17).

 

Sachais and colleagues retrospectively studied 34 FH patients treated with biweekly lipoprotein apheresis at the Hospital of the University of Pennsylvania (35). As expected, there was a marked reduction of LDL cholesterol level after apheresis and in some but not all patients there was a long-term reduction in their pre-apheresis LDL cholesterol levels. There was a marked decrease in cardiovascular events (3.2-fold decrease) defined as myocardial infarction, stroke, transient ischemic attack or rupture of aortic aneurysm. Similarly, there was also a 20-fold decrease in the need for cardiovascular interventions (coronary artery bypass surgery, carotid endarterectomy, and coronary artery angioplasty or stent placement).

 

Berent et al in an observational study of 30 patients reported that the incidence of cardiovascular disease 2 years after initiating apheresis compared to the 2 years prior to was reduced by 78% (36).

 

STUDIES FOCUSING ON LDL CHOLESTEROL AND LP(a)

 

In a single center study Koziolek and colleagues determined the incidence of major cardiovascular events in 38 patients who were treated during a 20 year period (37). LDL cholesterol and Lp(a) were reduced by approximately 60%. Major cardiovascular events were decreased from 7.02% events per patient per year at the start of lipid apheresis to 1.17% during lipid apheresis. Similarly, the rate of myocardial revascularization decreased from 22.8% to 3.8% per patient per year.

 

A multicenter study by von Dryander and colleagues examined the occurrence of cardiovascular events before apheresis and during apheresis in three groups defined by their lipid patterns at the start of an apheresis treatment: Group 1 (LDL-C ≥ 133mg/dl and Lp(a) ≤ 60 mg/dl; n = 35), Group 2 (LDL-C ≤ 133mg/dl and Lp(a) ≥ 60 mg/dl n = 37), and Group 3 (LDL-C ≥ 133mg/dl and Lp(a) ≥ 60 mg/dl; n = 15) (38). LDL cholesterol and Lp(a) levels were decreased by 55-70% by lipoprotein apheresis. Comparisons of the two years before the start of apheresis treatment with the first two years of apheresis treatment revealed the following reductions in the rates of cardiovascular events: Group 1- 54%; Group 2- 83%; Group 3- 83.5%.

 

In a single center study, Heigl and colleagues examined the effect of lipoprotein apheresis on cardiovascular events in 118 patients with either severe hypercholesterolemia or isolated increases in Lp(a) (39). Medium interval between the first cardiovascular event and apheresis treatment was 6.4 ± 5.6 years and the average apheresis treatment period was 6.8 ± 4.9 years. In patients with severe hypercholesterolemia (n=83) baseline LDL cholesterol levels were 176mg/dl and decreased by 67% following apheresis leading to an interval mean value of 120mg/dl. In patients with isolated elevations in Lp(a) (n=35), the baseline Lp(a) was 127mg/dl and decreased by 67% following apheresis leading to an interval mean value of 60mg/dl. After the initiation of lipoprotein apheresis, the annual rate of major cardiovascular events decreased by 80% (p<0.0001). Subgroup analysis showed a 73.7% decrease in patients with severe hypercholesterolemia (p<0.0001) and a 90.4% decrease in patients with isolated elevated Lp(a) levels (p< 0.0001).

 

Jaeger and colleagues in a longitudinal, multicenter, cohort study determined the effect of lipoprotein apheresis on major coronary events in 120 patients on maximal medical therapy with elevated LDL cholesterol (127mg/dl) and Lp(a) levels (>2.14micromol/l) (40). The mean duration of lipid-lowering therapy alone was 5.6 years and that of apheresis was 5.0 years. Median Lp(a) concentration was reduced from 4.00 micromol/l to 1.07 micromol/l (73% decrease) with apheresis treatment (P<0.0001) while LDL cholesterol levels decreased from 127mg/dl to 86mg/dl. Most importantly, major cardiovascular events were reduced by 86% during the lipoprotein apheresis phase (Annual rate 1.056 per patient during the pre-apheresis phase vs. 0.144 per patient during the apheresis phase; p < 0.0001).

 

In a review of data from the German Lipoprotein Apheresis Registry, the effect of lipoprotein apheresis in 991 patients was described (41). As expected, lipoprotein apheresis reduced both LDL cholesterol and Lp(a) levels by greater than 60%. Moreover, there was a 90% decrease in major adverse coronary events as well as a decrease in major adverse non-coronary events by 69 %. An update from the German Lipoprotein Apheresis Registry with 2028 reported similar results (42). Similarly, data from the United Kingdom registry reported a reduction in LDL cholesterol and Lp(a) of approximately 40% with a 62.5% reduction in major adverse cardiovascular events between the 2 years prior to, and the first 2 years following introduction of lipoprotein apheresis (43).

 

STUDIES FOCUSING ON LP(a)

 

Rosada and colleagues compared the occurrence of cardiovascular events in 37 patients with elevated Lp(a) levels (112mg/dl) and normal LDL cholesterol levels (84mg/dl) before the initiation of apheresis and during apheresis treatment (44). As expected, lipoprotein apheresis resulted in a marked decrease in LDL cholesterol levels (-60%) and Lp(a) levels (-68%). Event-free survival rate after 1 year in the pre-apheresis period was 38% vs. 75% during the apheresis period (P < 0.0001). These results suggest that lowering LDL cholesterol and Lp(a) levels in patients with normal LDL levels and elevated Lp(a) levels by lipoprotein apheresis reduces the number of cardiovascular events.

 

Leebmann, Roeseler and colleagues carried out a five year prospective observational multicenter study that compared cardiovascular events before and after lipoprotein apheresis in 170 patients with normal LDL cholesterol levels (99mg/dl) and elevated Lp(a) levels (108mg/dl) (45,46). As expected, apheresis reduced Lp(a) levels 68%. Moreover, there was a significant decline of the mean annual cardiovascular event rate from 0.58±0.53 2 years before initiating lipoprotein apheresis to 0.11±0.15 thereafter (P<0.0001). These results further support the hypothesis that lowering Lp(a) levels by apheresis in patients with elevated Lp(a) levels and reasonable LDL cholesterol levels will decrease cardiovascular events.

 

Grob et al studied 59 patients with elevated Lp(a) levels who were treated with lipoprotein apheresis (47). Lp(a) levels were acutely reduced by approximately 70% by apheresis and pre-apheresis Lp(a) levels were decreased by 22.8% compared to baseline. Moreover, cardiovascular events were reduced by approximately 83% during lipoprotein apheresis. Recently, Bigazzi reported in 23 patients with elevated Lp(a) levels and LDL cholesterol levels less than 100mg/dl that lipoprotein apheresis also resulted in a 74% reduction in cardiovascular events during apheresis compared to prior to apheresis (48).

 

Moriarty et al compared cardiovascular events pre and on lipoprotein apheresis in 14 patients with a mean of LDL cholesterol 80mg/dl and Lp(a) level of 138mg/dl pre-lipoprotein apheresis (49). On lipoprotein apheresis LDL cholesterol decreased to 29mg/dl and Lp(a) to 51mg/dl. Notably there was a 94% reduction in major adverse cardiovascular events over a mean treatment period of 48 months.

 

Finally, in a small study Poller and colleagues determined the effect of lipoprotein apheresis in 10 patients with peripheral artery disease who had recently undergone a revascularization procedure and had isolated elevations in Lp(a) (Lp(a) 156mg/dl; LDL cholesterol 85mg/dl) (50). After 12 months it was noted that the ankle-brachial-index increased from 0.5 ± 0.2 to 0.9 ± 0.1 (P < 0.001), the mean pain level decreased from 7.0 ± 1.5 to 2.0 ± 0.8 (P < 0.001) as determined using the visual analog scale, and that walking distance increased from 87 ± 60 m to 313 ± 145 m (P < 0.001). Moreover, the frequency of revascularization procedures was decreased (35 revascularizations within the 12 months prior to initiating apheresis vs. 1 revascularization procedure after starting apheresis P<0.001).

 

While the results of these studies are impressive and demonstrate a consistent reduction in cardiovascular events with the initiation of lipoprotein apheresis in patients with elevations in LDL cholesterol and/or Lp(a) levels it should be recognized that these studies did not include control groups. The absence of a control group is a major limitation. The patients included in these studies were likely selected for treatment with lipoprotein apheresis because they were having progressive cardiovascular events. The decrease in cardiovascular events following the initiation of lipoprotein apheresis could simply represent “regression to the mean” rather than a beneficial effect of apheresis. The inclusion of matched controls who were not treated with lipoprotein apheresis would have increased the significance and the reliability of the above observations. Of course, whether it would be ethical to include such a control group is debatable.

 

Controlled Trials

 

STUDIES FOCUSING ON LDL CHOLESTEROL

 

Koga et al determined the effect of the combination of lipoprotein apheresis plus drug therapy in 2 patients with homozygous FH and 9 patients with heterozygous FH compared to 10 heterozygous FH patients maintained on medication only on carotid intima-media thickness over a greater than 5 year period (51). It should be noted that the medication only group was significantly older than the apheresis group. The annual rate of progression of mean maximum intima-media thickness in the common carotid artery was -0.0023+/-0.0246 mm year in heterozygous FH patients treated with LDL apheresis plus drugs while in heterozygote FH patients treated with drugs alone the mean change was +0.0251+/-0.0265 mm year. These results suggest that the long-term treatment with combined lipoprotein apheresis and drugs may delay the progression of the atherosclerotic process and prompt the stabilization of atheromatous plaque in severe FH patients. However, it should be recognized that this was a small non-randomized study and the lipoprotein apheresis plus medication group was not perfectly matched with the medication only group.

           

 

Nishimura and colleagues compared angiographic changes after 2.3 years in 25 patients with heterozygous FH treated with lipoprotein apheresis and lipid lowering drugs and 11 patients who declined apheresis and were treated only with drugs (52). The apheresis plus drug therapy group was very similar to the lipid lowering drug therapy group. During the trial LDL-cholesterol levels were 140 +/- 34 mg/dl in the apheresis group and 170 +/- 58 mg/dl in the control group (P < 0.05). The mean changes in minimal lumen diameter of lesions were +0.19 +/- 0.30 mm (improved) in the apheresis group (n = 76) and -0.44 +/- 0.40 mm (worsened) in the control group (n = 37) (P < 0.0001). When progression and regression were defined as a change in minimal lumen diameter of +/- 0.67 mm, in the apheresis group, two patients (8%) had progression, 19 (76%) remained unchanged and four (16%) demonstrated regression. In contrast, in the control group seven patients (64%) had progression and four (36%) stayed unchanged. The frequency of regression or no change was greater in the apheresis group than in the control group (P < 0.004). It should be recognized that this was not a randomized study and there may have been subtle differences between the two groups.

 

 

Mabuchi and colleagues described the effects of lipoprotein apheresis on coronary artery disease in 43 patients with heterozygous FH treated with cholesterol lowering drug therapy plus apheresis vs. 87 patients with heterozygous FH treated with drug therapy alone (53). The patients were not randomized and there were differences in smoking, baseline LDL levels, and percent of patients with coronary artery bypass surgery between the apheresis vs. the drug only group. In the patients treated with apheresis the decrease in LDL cholesterol was 58% (LDL cholesterol on treatment 122mg/dl) while in the drug only group the decrease in LDL cholesterol was 28% (LDL cholesterol on treatment 168mg/dl). Major cardiovascular events including nonfatal myocardial infarction, percutaneous transluminal coronary angioplasty, coronary artery bypass grafting, and death from coronary heart disease were 72% lower in the lipoprotein apheresis group (10%) compared to the drug therapy alone group (36%) (p=0.0088). The lack of randomization and differences in the treatment groups limit the conclusions of this study.

 

Matsuzaki et al determined the effect of lipoprotein apheresis for one year in 18 patients with heterozygous FH on minimal lumen diameter (MLD) measured by coronary angiogram and plaque area (PA) measured by intravascular ultrasound (IVUS) (54). All patients were offered lipoprotein apheresis therapy and 11 patients elected to be treated with medication plus apheresis and 7 patients elected medications alone. The two groups were similar. The apheresis group showed a 34.3% reduction in LDL cholesterol from 213 +/- 25 mg/dl to 140 +/- 27 mg/dl) after one-year. The medication alone group showed no change in LDL cholesterol levels (174mg/dl at baseline and 181mg/dl at one year). Analysis of minimal lumen diameter (MLD) by coronary angiogram revealed an increase in MLD in the apheresis group and a decrease in the medication only group (P=0.008). Analysis of plaque area (PA) by IVUS revealed a decrease in the apheresis group and an increase in the medication only group (p=0.017). Once again, the lack of randomization and the potential for subtle differences in the two groups limit the conclusions of this study.

 

While the four studies described above were not randomized controlled trials, they nevertheless suggest that lipoprotein apheresis has beneficial effects on the progression of atherosclerosis and the occurrence of cardiovascular events.

 

In a 2-year randomized trial by Kroon and colleagues 42 men with severe coronary atherosclerosis were randomized to simvastatin 40mg daily (n=21) or simvastatin 40mg daily plus lipoprotein apheresis (n=21) (55). Baseline LDL cholesterol levels were approximately 300mg/dl and were reduced by 47% in the simvastatin group and 63% in the simvastatin plus apheresis group. No significant differences in quantitative coronary angiographic end points were observed between the two groups. However, in the simvastatin plus apheresis group bicycle exercise testing revealed a 39% increase in the time to 0.1 mV ST-segment depression and the maximum level of ST depression decreased significantly by 0.07 mV versus no changes in the simvastatin only group. Moreover, regional myocardial perfusion improved in the LDL apheresis group and remained unchanged in the medication group (56). Additionally, mean intima-media thickness decreased by 0.05 +/- 0.34 mm in the apheresis group and increased by 0.06 +/- 0.38 mm in the simvastatin-only group (P < 0.001) while the number of patients with hemodynamically significant stenosis in the aorta-tibial vessels decreased from 9 to 7 in the apheresis group and increased from 6 to 13 in the simvastatin alone group (P = 0.002) (57). Thus, this study showed that apheresis resulted in functional improvements and a decrease in atherosclerosis in non-coronary vessels. Atherosclerosis in the coronary arteries was not improved by apheresis during this 2-year study. Nevertheless, this randomized trial demonstrates that lipoprotein apheresis has benefits in patients with marked elevations in LDL cholesterol levels.

 

STUDIES FOCUSING ON LP(a)

 

Ezhov and colleagues studied 30 patients who had coronary heart disease with Lp(a) levels ≥50 mg/dL and LDL cholesterol levels ≤ 100 mg/dL on chronic statin therapy (58). Subjects were allocated to treatment with weekly apheresis with an immunoadsorption column specific for Lp(a) ("Lp(a) Lipopak"(®), POCARD Ltd., Russia) plus atorvastatin (n=15) or atorvastatin monotherapy (n=15). As expected in the apheresis group Lp(a) level decreased by an average of 73 ± 12% to a mean of 29 ± 16 mg/dL while there was no significant change in the atorvastatin monotherapy group. Moreover, carotid intima-media thickness (CIMT) did not change in the atorvastatin alone group but in the apheresis group CIMT at 9 and 18 months decreased from baseline by -0.03 ± 0.09 mm (p = 0.05) and -0.07 ± 0.15 mm (p = 0.01), respectively. Additionally, clinical status was improved, with less angina in the apheresis group. This controlled trial demonstrates that lowering Lp(a) by apheresis has beneficial effects on atherosclerosis as determined by measuring CIMT.

 

In contrast, a study by Thompson and colleagues did not demonstrate a benefit of lowering Lp(a) by lipoprotein apheresis (59). In this trial patients with heterozygous FH were randomized to simvastatin 40mg daily plus apheresis (n=20) or simvastatin plus colestipol (n=19). LDL cholesterol levels were slightly lower in the apheresis group (125mg/dl vs. 133mg/dl, p= 0.03) while Lp(a) levels were reduced by 33% (14mg/dl vs. 21mg/dl, p=0.03). After a mean of 2.1 years there were no differences in quantitative coronary angiography between the two groups. The results of this study suggest no benefit to lowering Lp(a) levels. However, it should be noted that in this study the Lp(a) levels were not very high and therefore this study did not examine the effect of lowering Lp(a) levels in patients with elevated levels.

 

Finally, in a small study by Khan and colleagues randomized 20 patients with refractory angina and elevated Lp(a) >500 mg/L (normal <300 mg/L) and an LDL cholesterol level less than 156mg/dl (4.0 mmol/L), despite optimal lipid lowering drug therapy to lipoprotein apheresis or a sham procedure (60). The reported that total carotid wall volume, a marker of atherosclerosis, increased in the sham group but decreased in the lipoprotein apheresis group (P < 0.001 between groups) suggesting that apheresis reduces atherosclerotic burden.

 

Summary

 

In conclusion, while the studies described above are not perfect and do not definitively demonstrate that lipoprotein apheresis decreases cardiovascular events, the results of the lipoprotein apheresis studies coupled with the randomized studies of LDL cholesterol lowering with statins and other drugs makes it extremely likely that lowering LDL cholesterol levels with lipoprotein apheresis will reduce the risk of cardiovascular events (12). Whether lowering Lp(a) levels with lipoprotein apheresis is somewhat less certain, as to date no intervention to lower Lp(a) levels has been shown to reduce events. Nevertheless given the epidemiology data, genetic studies, basic science experiments, and animal experiments it is very likely that lowering elevated Lp(a) will have beneficial effects on cardiovascular outcomes in patients with high Lp(a) levels (61,62).

 

OTHER BENEFITS OF LIPOPROTEIN APHERESIS

 

Randomized controlled trials have shown that a single lipoprotein apheresis was beneficial in restoring hearing in patients with acute hearing loss (63,64). Additionally, lipoprotein apheresis has been shown to induce remission in approximately 50% of patients with drug-resistant nephrotic syndrome (1,65). The FDA has approved lipoprotein apheresis for new onset focal segmental glomerulosclerosis in pediatric patients who are resistant to standard treatment (1). A meta-analysis has also reported benefit in adult patients with focal segmental glomerulosclerosis (66)

 

SIDE EFFECTS AND CONTRAINDICATIONS

 

Lipoprotein apheresis in general is well tolerated. During apheresis a decrease in blood pressure may occur in some patients (2,3,8,16). Additionally, with long standing apheresis iron deficiency anemia may occur (67).

 

Lipoprotein apheresis using polyacrylate and dextran sulfate columns converts kininogen to bradykinin leading to marked increases in bradykinin levels (68). Angiotensin converting enzyme (ACE) inactivates bradykinin and therefore treatment with ACE inhibitors is contraindicated in patients receiving lipoprotein apheresis with polyacrylate or dextran sulfate as the resulting very high levels of bradykinin may lead to severe hypotension and an anaphylactoid reaction (2,3,68,69). However, in these patient’s angiotensin receptor blockers can be safely used.

 

CONCLUSION

 

Lipoprotein apheresis is a well-tolerated procedure that markedly lowers LDL cholesterol and Lp(a) levels in patients who do not obtain acceptable levels with maximal lifestyle and drug therapy. Studies strongly suggest that lipoprotein apheresis will decrease the progression of atherosclerosis and reduce cardiovascular events. Therefore, lipoprotein apheresis is a potential treatment in selected patients with drug resistant elevations in LDL cholesterol and/or Lp(a) levels. Studies have shown that lipoprotein apheresis safely reduces LDL cholesterol levels and xanthomas in children with homozygous Familial Hypercholesterolemia (21).

 

REFERENCES

 

  1. Moriarty PM. Lipoprotein apheresis: present and future uses. Curr Opin Lipidol 2015; 26:544-552
  2. Moriarty PM, Hemphill L. Lipoprotein Apheresis. Endocrinol Metab Clin North Am 2016; 45:39-54
  3. Stefanutti C, Thompson GR. Lipoprotein apheresis in the management of familial hypercholesterolaemia: historical perspective and recent advances. Curr Atheroscler Rep 2015; 17:465
  4. Waldmann E, Parhofer KG. Lipoprotein apheresis to treat elevated lipoprotein (a). J Lipid Res 2016; 57:1751-1757
  5. Schettler V, Neumann CL, Hulpke-Wette M, Hagenah GC, Schulz EG, Wieland E, German Apheresis Working G. Current view: indications for extracorporeal lipid apheresis treatment. Clin Res Cardiol Suppl 2012; 7:15-19
  6. Harada-Shiba M, Arai H, Oikawa S, Ohta T, Okada T, Okamura T, Nohara A, Bujo H, Yokote K, Wakatsuki A, Ishibashi S, Yamashita S. Guidelines for the management of familial hypercholesterolemia. J Atheroscler Thromb 2012; 19:1043-1060
  7. Jacobson TA, Ito MK, Maki KC, Orringer CE, Bays HE, Jones PH, McKenney JM, Grundy SM, Gill EA, Wild RA, Wilson DP, Brown WV. National Lipid Association recommendations for patient-centered management of dyslipidemia: part 1 - executive summary. J Clin Lipidol 2014; 8:473-488
  8. Julius U. Lipoprotein apheresis in the management of severe hypercholesterolemia and of elevation of lipoprotein(a): current perspectives and patient selection. Med Devices (Auckl) 2016; 9:349-360
  9. Graham DF, Raal FJ. Management of familial hypercholesterolemia in pregnancy. Curr Opin Lipidol 2021; 32:370-377
  10. Taylan C, Driemeyer J, Schmitt CP, Pape L, Buscher R, Galiano M, Konig J, Schurfeld C, Spitthover R, Versen A, Koziolek M, Marsen TA, Stein H, Schaefer JR, Heibges A, Klingel R, Oh J, Weber LT, Klaus G. Cardiovascular Outcome of Pediatric Patients With Bi-Allelic (Homozygous) Familial Hypercholesterolemia Before and After Initiation of Multimodal Lipid Lowering Therapy Including Lipoprotein Apheresis. Am J Cardiol2020; 136:38-48
  11. Luirink IK, Hutten BA, Greber-Platzer S, Kolovou GD, Dann EJ, de Ferranti SD, Taylan C, Bruckert E, Saheb S, Oh J, Driemeyer J, Farnier M, Pape L, Schmitt CP, Novoa FJ, Maeser M, Masana L, Shahrani A, Wiegman A, Groothoff JW. Practice of lipoprotein apheresis and short-term efficacy in children with homozygous familial hypercholesterolemia: Data from an international registry. Atherosclerosis 2020; 299:24-31
  12. Feingold KR. Cholesterol Lowering Drugs. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Hofland J, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2021.
  13. Baum SJ, Sampietro T, Datta D, Moriarty PM, Knusel B, Schneider J, Somaratne R, Kurtz C, Hohenstein B. Effect of evolocumab on lipoprotein apheresis requirement and lipid levels: Results of the randomized, controlled, open-label DE LAVAL study. J Clin Lipidol 2019; 13:901-909 e903
  14. Tsimikas S, Viney NJ, Hughes SG, Singleton W, Graham MJ, Baker BF, Burkey JL, Yang Q, Marcovina SM, Geary RS, Crooke RM, Witztum JL. Antisense therapy targeting apolipoprotein(a): a randomised, double-blind, placebo-controlled phase 1 study. Lancet 2015; 386:1472-1483
  15. Viney NJ, van Capelleveen JC, Geary RS, Xia S, Tami JA, Yu RZ, Marcovina SM, Hughes SG, Graham MJ, Crooke RM, Crooke ST, Witztum JL, Stroes ES, Tsimikas S. Antisense oligonucleotides targeting apolipoprotein(a) in people with raised lipoprotein(a): two randomised, double-blind, placebo-controlled, dose-ranging trials. Lancet 2016; 388:2239-2253
  16. Wang A, Richhariya A, Gandra SR, Calimlim B, Kim L, Quek RG, Nordyke RJ, Toth PP. Systematic Review of Low-Density Lipoprotein Cholesterol Apheresis for the Treatment of Familial Hypercholesterolemia. J Am Heart Assoc 2016; 5
  17. Schmaldienst S, Banyai S, Stulnig TM, Heinz G, Jansen M, Horl WH, Derfler K. Prospective randomised cross-over comparison of three LDL-apheresis systems in statin pretreated patients with familial hypercholesterolaemia. Atherosclerosis 2000; 151:493-499
  18. Schaumann D, Welch-Wichary M, Voss A, Schmidt H, Olbricht CJ. Prospective cross-over comparisons of three low-density lipoprotein (LDL)-apheresis methods in patients with familial hypercholesterolaemia. Eur J Clin Invest 1996; 26:1033-1038
  19. Kroon AA, van't Hof MA, Demacker PN, Stalenhoef AF. The rebound of lipoproteins after LDL-apheresis. Kinetics and estimation of mean lipoprotein levels. Atherosclerosis 2000; 152:519-526
  20. Walji S NC, Thompson GR. Lipoprotein Apheresis for the Treatment of Familial Hypercholesterolemia. Clin Lipidology 2013; 8:573-586
  21. Luirink IK, Determeijer J, Hutten BA, Wiegman A, Bruckert E, Schmitt CP, Groothoff JW. Efficacy and safety of lipoprotein apheresis in children with homozygous familial hypercholesterolemia: A systematic review. J Clin Lipidol 2019; 13:31-39
  22. Click B, Ketchum AM, Turner R, Whitcomb DC, Papachristou GI, Yadav D. The role of apheresis in hypertriglyceridemia-induced acute pancreatitis: A systematic review. Pancreatology 2015; 15:313-320
  23. Ramirez-Bueno A, Salazar-Ramirez C, Cota-Delgado F, de la Torre-Prados MV, Valdivielso P. Plasmapheresis as treatment for hyperlipidemic pancreatitis. Eur J Intern Med 2014; 25:160-163
  24. Julius U, Milton M, Stoellner D, Rader D, Gordon B, Polk D, Waldmann E, Parhofer KG, Moriarty PM. Effects of lipoprotein apheresis on PCSK9 levels. Atheroscler Suppl 2015; 18:180-186
  25. Thompson GR, Miller JP, Breslow JL. Improved survival of patients with homozygous familial hypercholesterolaemia treated with plasma exchange. Br Med J (Clin Res Ed) 1985; 291:1671-1673
  26. Keller C. LDL-apheresis in homozygous LDL-receptor-defective familial hypercholesterolemia: the Munich experience. Atheroscler Suppl 2009; 10:21-26
  27. Thompson GR, Myant NB, Kilpatrick D, Oakley CM, Raphael MJ, Steiner RE. Assessment of long-term plasma exchange for familial hypercholesterolaemia. Br Heart J 1980; 43:680-688
  28. Tatami R, Inoue N, Itoh H, Kishino B, Koga N, Nakashima Y, Nishide T, Okamura K, Saito Y, Teramoto T, et al. Regression of coronary atherosclerosis by combined LDL-apheresis and lipid-lowering drug therapy in patients with familial hypercholesterolemia: a multicenter study. The LARS Investigators. Atherosclerosis 1992; 95:1-13
  29. Schuff-Werner P, Gohlke H, Bartmann U, Baggio G, Corti MC, Dinsenbacher A, Eisenhauer T, Grutzmacher P, Keller C, Kettner U, et al. The HELP-LDL-apheresis multicentre study, an angiographically assessed trial on the role of LDL-apheresis in the secondary prevention of coronary heart disease. II. Final evaluation of the effect of regular treatment on LDL-cholesterol plasma concentrations and the course of coronary heart disease. The HELP-Study Group. Heparin-induced extra-corporeal LDL-precipitation. Eur J Clin Invest 1994; 24:724-732
  30. Waidner T, Franzen D, Voelker W, Ritter M, Borberg H, Hombach V, Hopp HW. The effect of LDL apheresis on progression of coronary artery disease in patients with familial hypercholesterolemia. Results of a multicenter LDL apheresis study. Clin Investig 1994; 72:858-863
  31. Richter WO, Donner MG, Hofling B, Schwandt P. Long-term effect of low-density lipoprotein apheresis on plasma lipoproteins and coronary heart disease in native vessels and coronary bypass in severe heterozygous familial hypercholesterolemia. Metabolism 1998; 47:863-868
  32. Stefanutti C, D'Alessandri G, Russi G, De Silvestro G, Zenti MG, Marson P, Belotherkovsky D, Vivenzio A, Di Giacomo S. Treatment of symptomatic HyperLp(a)lipoproteinemia with LDL-apheresis: a multicentre study. Atheroscler Suppl 2009; 10:89-94
  33. Safarova MS, Nugent AK, Gorby L, Dutton JA, Thompson WJ, Moriarty PM. Effect of Lipoprotein Apheresis on Progression of Carotid Intima-Media Thickness in Patients with Severe Hypercholesterolemia. Am J Cardiol2022; 177:22-27
  34. Gordon BR, Kelsey SF, Dau PC, Gotto AM, Jr., Graham K, Illingworth DR, Isaacsohn J, Jones PH, Leitman SF, Saal SD, Stein EA, Stern TN, Troendle A, Zwiener RJ. Long-term effects of low-density lipoprotein apheresis using an automated dextran sulfate cellulose adsorption system. Liposorber Study Group. Am J Cardiol 1998; 81:407-411
  35. Sachais BS, Katz J, Ross J, Rader DJ. Long-term effects of LDL apheresis in patients with severe hypercholesterolemia. J Clin Apher 2005; 20:252-255
  36. Berent T, Derfler K, Berent R, Sinzinger H. Lipoprotein apheresis in Austria - Reduction of cardiovascular events by regular lipoprotein apheresis treatment. Atheroscler Suppl 2019; 40:8-11
  37. Koziolek MJ, Hennig U, Zapf A, Bramlage C, Grupp C, Armstrong VW, Strutz F, Muller GA. Retrospective analysis of long-term lipid apheresis at a single center. Ther Apher Dial 2010; 14:143-152
  38. von Dryander M, Fischer S, Passauer J, Muller G, Bornstein SR, Julius U. Differences in the atherogenic risk of patients treated by lipoprotein apheresis according to their lipid pattern. Atheroscler Suppl 2013; 14:39-44
  39. Heigl F, Hettich R, Lotz N, Reeg H, Pflederer T, Osterkorn D, Osterkorn K, Klingel R. Efficacy, safety, and tolerability of long-term lipoprotein apheresis in patients with LDL- or Lp(a) hyperlipoproteinemia: Findings gathered from more than 36,000 treatments at one center in Germany. Atheroscler Suppl 2015; 18:154-162
  40. Jaeger BR, Richter Y, Nagel D, Heigl F, Vogt A, Roeseler E, Parhofer K, Ramlow W, Koch M, Utermann G, Labarrere CA, Seidel D, Group of Clinical I. Longitudinal cohort study on the effectiveness of lipid apheresis treatment to reduce high lipoprotein(a) levels and prevent major adverse coronary events. Nat Clin Pract Cardiovasc Med 2009; 6:229-239
  41. Schettler VJ, Neumann CL, Peter C, Zimmermann T, Julius U, Roeseler E, Heigl F, German Apheresis Working G. Impact of the German Lipoprotein Apheresis Registry (DLAR) on therapeutic options to reduce increased Lp(a) levels. Clin Res Cardiol Suppl 2015; 10:14-20
  42. Schettler VJJ, Peter C, Zimmermann T, Julius U, Roeseler E, Schlieper G, Heigl F, Grutzmacher P, Lohlein I, Klingel R, Hohenstein B, Ramlow W, Vogt A, Scientific Board of GftGAWG. The German Lipoprotein Apheresis Registry-Summary of the ninth annual report. Ther Apher Dial 2022; 26 Suppl 1:81-88
  43. Pottle A, Thompson G, Barbir M, Bayly G, Cegla J, Cramb R, Dawson T, Eatough R, Kale V, Neuwirth C, Nicholson K, Payne J, Scott J, Soran H, Walji S, Watkins S, Weedon H, Nath Datta DB. Lipoprotein apheresis efficacy, challenges and outcomes: A descriptive analysis from the UK Lipoprotein Apheresis Registry, 1989-2017. Atherosclerosis 2019; 290:44-51
  44. Rosada A, Kassner U, Vogt A, Willhauck M, Parhofer K, Steinhagen-Thiessen E. Does regular lipid apheresis in patients with isolated elevated lipoprotein(a) levels reduce the incidence of cardiovascular events? Artif Organs2014; 38:135-141
  45. Leebmann J, Roeseler E, Julius U, Heigl F, Spitthoever R, Heutling D, Breitenberger P, Maerz W, Lehmacher W, Heibges A, Klingel R, ProLiFe Study G. Lipoprotein apheresis in patients with maximally tolerated lipid-lowering therapy, lipoprotein(a)-hyperlipoproteinemia, and progressive cardiovascular disease: prospective observational multicenter study. Circulation 2013; 128:2567-2576
  46. Roeseler E, Julius U, Heigl F, Spitthoever R, Heutling D, Breitenberger P, Leebmann J, Lehmacher W, Kamstrup PR, Nordestgaard BG, Maerz W, Noureen A, Schmidt K, Kronenberg F, Heibges A, Klingel R, ProLiFe-Study G. Lipoprotein Apheresis for Lipoprotein(a)-Associated Cardiovascular Disease: Prospective 5 Years of Follow-Up and Apolipoprotein(a) Characterization. Arterioscler Thromb Vasc Biol 2016; 36:2019-2027
  47. Gross E, Hohenstein B, Julius U. Effects of Lipoprotein apheresis on the Lipoprotein(a) levels in the long run. Atheroscler Suppl 2015; 18:226-232
  48. Bigazzi F, Sbrana F, Berretti D, Maria Grazia Z, Zambon S, Fabris A, Fonda M, Vigna GB, D'Alessandri G, Passalacqua S, Dal Pino B, Pianelli M, Luciani R, Ripoli A, Rafanelli D, Manzato E, Cattin L, Sampietro T. Reduced incidence of cardiovascular events in hyper-Lp(a) patients on lipoprotein apheresis. The G.I.L.A. (Gruppo Interdisciplinare Aferesi Lipoproteica) pilot study. Transfus Apher Sci 2018; 57:661-664
  49. Moriarty PM, Gray JV, Gorby LK. Lipoprotein apheresis for lipoprotein(a) and cardiovascular disease. J Clin Lipidol 2019; 13:894-900
  50. Poller WC, Dreger H, Morgera S, Berger A, Flessenkamper I, Enke-Melzer K. Lipoprotein apheresis in patients with peripheral artery disease and hyperlipoproteinemia(a). Atheroscler Suppl 2015; 18:187-193
  51. Koga N, Watanabe K, Kurashige Y, Sato T, Hiroki T. Long-term effects of LDL apheresis on carotid arterial atherosclerosis in familial hypercholesterolaemic patients. J Intern Med 1999; 246:35-43
  52. Nishimura S, Sekiguchi M, Kano T, Ishiwata S, Nagasaki F, Nishide T, Okimoto T, Kutsumi Y, Kuwabara Y, Takatsu F, Nishikawa H, Daida H, Yamaguchi H. Effects of intensive lipid lowering by low-density lipoprotein apheresis on regression of coronary atherosclerosis in patients with familial hypercholesterolemia: Japan Low-density Lipoprotein Apheresis Coronary Atherosclerosis Prospective Study (L-CAPS). Atherosclerosis 1999; 144:409-417
  53. Mabuchi H, Koizumi J, Shimizu M, Kajinami K, Miyamoto S, Ueda K, Takegoshi T. Long-term efficacy of low-density lipoprotein apheresis on coronary heart disease in familial hypercholesterolemia. Hokuriku-FH-LDL-Apheresis Study Group. Am J Cardiol 1998; 82:1489-1495
  54. Matsuzaki M, Hiramori K, Imaizumi T, Kitabatake A, Hishida H, Nomura M, Fujii T, Sakuma I, Fukami K, Honda T, Ogawa H, Yamagishi M. Intravascular ultrasound evaluation of coronary plaque regression by low density lipoprotein-apheresis in familial hypercholesterolemia: the Low Density Lipoprotein-Apheresis Coronary Morphology and Reserve Trial (LACMART). J Am Coll Cardiol 2002; 40:220-227
  55. Kroon AA, Aengevaeren WR, van der Werf T, Uijen GJ, Reiber JH, Bruschke AV, Stalenhoef AF. LDL-Apheresis Atherosclerosis Regression Study (LAARS). Effect of aggressive versus conventional lipid lowering treatment on coronary atherosclerosis. Circulation 1996; 93:1826-1835
  56. Aengevaeren WR, Kroon AA, Stalenhoef AF, Uijen GJ, van der Werf T. Low density lipoprotein apheresis improves regional myocardial perfusion in patients with hypercholesterolemia and extensive coronary artery disease. LDL-Apheresis Atherosclerosis Regression Study (LAARS). J Am Coll Cardiol 1996; 28:1696-1704
  57. Kroon AA, van Asten WN, Stalenhoef AF. Effect of apheresis of low-density lipoprotein on peripheral vascular disease in hypercholesterolemic patients with coronary artery disease. Ann Intern Med 1996; 125:945-954
  58. Ezhov MV, Safarova MS, Afanasieva OI, Pogorelova OA, Tripoten MI, Adamova IY, Konovalov GA, Balakhonova TV, Pokrovsky SN. Specific Lipoprotein(a) apheresis attenuates progression of carotid intima-media thickness in coronary heart disease patients with high lipoprotein(a) levels. Atheroscler Suppl 2015; 18:163-169
  59. Thompson GR, Maher VM, Matthews S, Kitano Y, Neuwirth C, Shortt MB, Davies G, Rees A, Mir A, Prescott RJ, et al. Familial Hypercholesterolaemia Regression Study: a randomised trial of low-density-lipoprotein apheresis. Lancet 1995; 345:811-816
  60. Khan TZ, Hsu LY, Arai AE, Rhodes S, Pottle A, Wage R, Banya W, Gatehouse PD, Giri S, Collins P, Pennell DJ, Barbir M. Apheresis as novel treatment for refractory angina with raised lipoprotein(a): a randomized controlled cross-over trial. Eur Heart J 2017; 38:1561-1569
  61. Nordestgaard BG, Langsted A. Lipoprotein (a) as a cause of cardiovascular disease: insights from epidemiology, genetics, and biology. J Lipid Res 2016; 57:1953-1975
  62. Hung MY, Tsimikas S. What is the ultimate test that lowering lipoprotein(a) is beneficial for cardiovascular disease and aortic stenosis? Curr Opin Lipidol 2014; 25:423-430
  63. Suckfull M, Hearing Loss Study G. Fibrinogen and LDL apheresis in treatment of sudden hearing loss: a randomised multicentre trial. Lancet 2002; 360:1811-1817
  64. Bianchin G, Russi G, Romano N, Fioravanti P. Treatment with HELP-apheresis in patients suffering from sudden sensorineural hearing loss: a prospective, randomized, controlled study. Laryngoscope 2010; 120:800-807
  65. Muso E, Mune M, Hirano T, Hattori M, Kimura K, Watanabe T, Yokoyama H, Sato H, Uchida S, Wada T, Shoji T, Takemura T, Yuzawa Y, Ogahara S, Sugiyama S, Iino Y, Sakai S, Ogura Y, Yukawa S, Nishizawa Y, Yorioka N, Imai E, Matsuo S, Saito T. A Prospective Observational Survey on the Long-Term Effect of LDL Apheresis on Drug-Resistant Nephrotic Syndrome. Nephron Extra 2015; 5:58-66
  66. Miao J, Krisanapan P, Tangpanithandee S, Thongprayoon C, Mao MA, Cheungpasitporn W. Efficacy of extracorporeal plasma therapy for adult native kidney patients with Primary FSGS: a Systematic review. Ren Fail 2023; 45:2176694
  67. Schatz U, Arneth B, Siegert G, Siegels D, Fischer S, Julius U, Bornstein SR. Iron deficiency and its management in patients undergoing lipoprotein apheresis. Comparison of two parenteral iron formulations. Atheroscler Suppl 2013; 14:115-122
  68. Krieter DH, Steinke J, Kerkhoff M, Fink E, Lemke HD, Zingler C, Muller GA, Schuff-Werner P. Contact activation in low-density lipoprotein apheresis systems. Artif Organs 2005; 29:47-52
  69. Koga N, Nagano T, Sato T, Kagasawa K. Anaphylactoid reactions and bradykinin generation in patients treated with LDL-apheresis and an ACE inhibitor. ASAIO J 1993; 39:M288-291

Insulin – Pharmacology, Therapeutic Regimens And Principles Of Intensive Insulin Therapy

ABSTRACT

 

Since the introduction of insulin analogs in 1996, insulin therapy options for patients with type 1 and type 2 diabetes have expanded. Insulin therapies are now able to more closely mimic physiologic insulin secretion and thus achievebetter glycemic control in patients with diabetes. This chapter reviews the pharmacology of available insulins, types of insulin regimens, and principles of dosage selection and adjustment, and provides an overview of insulin pump therapy.

 

INTRODUCTION

 

In 1922, Canadian researchers were the first to demonstrate a physiologic response to injected animal insulin in a patient with type 1 diabetes. Insulin was the first protein to be fully sequenced. The insulin molecule consists of 51 amino acids arranged in two chains, an A chain (21 amino acids) and B chain (30 amino acids) that are linked by two disulfide bonds (1) (Figure 1). Proinsulin is the insulin precursor that is transported to the Golgi apparatus of the beta cell where it is processed and packaged into granules. Proinsulin, a single-chain 86 amino acid peptide, is cleaved into insulin and C-peptide (a connecting peptide); both are secreted in equimolar portions from the beta cell upon stimulation from glucose and other insulin secretagogues. While C-peptide has no known physiologic function, it can be measured to provide an estimate of endogenous insulin secretion.

 

Figure 1. Insulin Structure.

SOURCES OF INSULIN

 

With the availability of human insulin by recombinant DNA technology in the 1980s, use of animal insulin declined dramatically. Beef insulin, beef-pork, and pork insulin are no longer commercially available in the United States. The United States FDA may allow for personal importation of beef or pork insulin from a foreign country if a patient cannot be treated with human insulin (2). Beef insulin differs from human insulin by 3 amino acids and pork insulin differs by one amino acid (2).

 

Currently, in the United States, insulins used are either human insulin and/or analogs of human insulin. The recombinant DNA technique for producing insulin for commercial use involves insertion of the human proinsulin gene into either Saccharomyces cerevisiae (baker’s yeast) or a non-pathogenic laboratory strain of Escherichia coli (E coli) which serve as the production organism. Human insulin is then isolated and purified (3–11) .

 

INSULIN ANALOGS

 

Recombinant DNA technology has allowed for the development and production of analogs to human insulin. With analogs, the insulin molecule structure is modified slightly to alter the pharmacokinetic properties of insulin, primarily affecting the absorption of the drug from the subcutaneous tissue. The B26-B30 region of the insulin molecule is not critical for insulin receptor recognition and it is in this region that amino acids are generally substituted (12).

 

Thus, the insulin analogs are still recognized by and bind to the insulin receptor. The structures of three rapid-acting insulin analogs are shown in Figure 2 (insulin aspart, lispro and glulisine) and the structures of three long-actinginsulin analogs are shown in Figure 3 (insulin glargine, detemir, and degludec).

 

Figure 2. Insulin Aspart, Glulisine and Lispro Structures.

Figure 3. Insulin Glargine and Detemir Structures.

In vitro studies have demonstrated the mitogenic effects of insulin at high concentrations, as well as carcinogeniceffects of insulin binding to the insulin like growth factor-1 (IGF-1) receptor,

 

suggesting that hyperinsulinemia may promote tumorigenesis. Subcutaneously administered insulin bypasses the usual 80% hepatic first pass clearance of pancreatic islet cell-secreted insulin, and therefore contributes to systemic hyperinsulinemia in insulin-treated patients with diabetes (13). Because insulin analogs are modified human insulin, the safety and efficacy profiles of these insulins have been compared to human insulin (12) . Insulin and IGF-1 receptor binding affinities, and the metabolic and mitogenic potencies of insulin analogs relative to human insulin have been assessed. Insulin lispro and aspart are similar to human insulin on all of the above parameters, except insulin lispro was found to be 1.5-fold more potent in binding to the IGF-1 receptor compared to human insulin. Insulin glargine was found to have a 6- to 8-fold increase in mitogenic potency and IGF- 1 receptor affinity compared to human insulin. However, glargine is rapidly degraded to metabolites. The predominant metabolite M1 has been shown to have a 0.4-fold binding affinity to the IGF-1 receptor compared with human insulin (14) . In human studies, meta-analyses comparing exogenous insulin to non-insulin antihyperglycemic therapies have shown associations of insulin with several cancers (15,16) . However, there are inherent limitations to such analyses. A review of large epidemiologic studies did not find evidence of an increased risk of malignancy among glargine-treated patients when compared with other insulin therapies (14). Observational studies with up to 7 years of follow up have also not shown an association of cancer with insulin glargine or detemir use (17).

 

Insulin detemir was found to be more than 5-fold less potent than human insulin in binding to IGF-1 (12). An in vitro study showed that insulin degludec had a low IGF-1 receptor binding affinity compared to human insulin (18) . Thelong-term clinical significance of differences in IGF- 1 binding among available insulins is not known.

 

IMMUNOGENICITY

 

Because pork and beef insulin differ from human insulin by 1 and 3 amino acids respectively, they are more immunogenic than exogenous human insulin. Older formulations of insulin were less pure, containing islet-cell peptides, proinsulin, C-peptide, pancreatic polypeptides, glucagon, and somatostatin, which contributed to the immunogenicity of insulin (19). Components of insulin preparations (e.g., zinc, protamine) and subcutaneous insulin aggregates are also thought to contribute to antibody formation (19). Because of the availability of human insulin and the increased potential for animal source insulin to be immunogenic, animal source insulins are no longer availablein the United States.

 

Rare hypersensitivity responses to insulin can be immediate-type, local or systemic IgE- mediated reactions (19) . Patients who experience a true allergic reaction to insulin have typically received insulin in the past, and experience the reaction after insulin is restarted. Delayed, IgG-mediated allergic reactions also develop with animal insulins (19). Insulin therapy can rarely result in the production of insulin antibodies of the IgG class, which inactivate insulin.Immunological insulin resistance can develop in patients with very high titers of IgG- antibodies.

 

Lipodystrophy resulting from insulin injections refers to two conditions: lipoatrophy and lipohypertrophy. Lipoatrophy is an immune-mediated condition resulting in loss of fat at insulin injection sites (19) and occurs rarely with purified human insulins. Treatment for patients who developed lipoatrophy due to animal insulin use was injection of human insulin into the atrophied site. Lipohypertrophy is a common, non-immunological side effect of insulin resulting frominsulin’s trophic effects following repeated injections of insulin into the same subcutaneous site (20) . Lipohypertrophy can delay the absorption of insulin and therefore it is best if patients do not continue to administer insulin in these locations.

 

CONCENTRATION

 

In the United States, all insulins are available in the concentration of 100 units/ml (denoted as U-100). Insulin syringes are designed to accommodate this concentration of insulin. Regular human insulin (Humulin R, Lilly) is available in a more concentrated insulin, U-500 (500 units/ml), and is used primarily in cases of marked insulin resistance, when large doses of insulin (generally > 200 units per day) are required. Extreme caution must be taken as each marked unit on a U-100 syringe will deliver 5 units of insulin. However, syringes specific to U- 500 insulin are available, and U-500 insulin is also available for administration via a pen device. For both the syringe and pen specific to U-500 insulin, the units, not the volume, of insulin are marked. Insulin glargine is also available in a U-300 concentration, delivering 300 units/ml, and insulin degludec and insulin lispro are available in U-200 concentrations that deliver 200 units/mL. Both U-300 and U-200 insulin are only available in pen devices, and for both U-300 and U-200, the dose of insulin a patient dials into the pen device is in units and not in mL.

 

Outside the United States, a less concentrated insulin preparation, U-40, (40 units/ml) is still available and sometimes used, although this has become uncommon (21). Specific U-40 syringes are used with this insulin. It is important thatpatients traveling from one country to the next be aware of the concentration of insulin they use and that the appropriate syringe is used.

 

PHYSICAL AND CHEMICAL PROPERTIES

 

Regular human insulin is crystalline zinc insulin dissolved in a clear solution. It may be administered by any parenteral route: subcutaneous, intramuscular, or intravenous. Insulin aspart, glulisine and lispro are also soluble crystalline zinc insulin, but are intended for subcutaneous (SQ) injection. When administered intravenously, the action of theserapid-acting insulin analogs is identical to that of regular insulin. NPH, or neutral protamine Hagedorn, is a suspension of regular insulin complexed with protamine that delays its absorption. Insulin suspensions should not be administered intravenously. All insulins, except insulin glargine, are formulated to a neutral pH.

 

Long-acting insulin glargine is a soluble, clear insulin, with a pH of 4.0 which affects its SQ absorption characteristics, discussed further in the pharmacokinetics section. Insulin glargine should not be mixed with other insulins, and should only be administered subcutaneously (8). Insulin detemir is an insulin analog coupled to an 18-chain fatty acid that binds to albumin in the SQ tissue. This results in delayed absorption and a prolonged duration of action. Insulin degludec is an ultra-long insulin analog that breaks down into monomers by dissociating from zinc molecules after administration (22). Insulins detemir and degludec should also not be mixed with other insulins and are intended only for subcutaneous use (5,7).

 

PHARMACOKINETICS

 

Absorption

 

Insulin administered via SQ injection is absorbed into the bloodstream, and the lymphatic system also plays a role intransport (23). The absorption of human insulin into the bloodstream after SQ absorption is the rate limiting stepof insulin activity. This absorption is inconsistent with the coefficients of variation of T50% (time for 50% of the insulin dose to be absorbed) varying ~ 15% within an individual and 30% between patients (24). Most of this variability of insulin absorption is correlated to blood flow differences at the various sites of injection (abdomen, deltoid, gluteus, and thigh) (25). For regular insulin, the impact of this is a more than 2 times faster rate of absorption from theabdomen than the thigh (25). The clinical significance of this is that patients should avoid random use of different body regions for their injections. For example, if a patient prefers to use their thigh for a noontime injection, this site should be used consistently for this injection. The abdomen is the preferred site of injection because it is the least susceptible to factors affecting insulin absorption (see Table 1). Insulin aspart, glulisine and lispro appear to have lessday-to-day variation in absorption rates and also less absorption variation from the different body regions (3,9,10,26). Insulin glargine’s pharmacokinetic profile is similar after abdominal, deltoid or thigh SQ administration (8). Similarly, the glucose-lowering effect of insulin degludec has not been found to vary between abdominal, upper arm, or thigh SQ sites (27) .

 

Factors that alter insulin absorption do so mostly by changing local blood flow in the SQ tissue. Factors thatincrease SQ blood flow increase the absorption rate. Table 1 lists factors that affect insulin absorption.

 

Table 1. Factors Affecting Insulin Absorption (12,28)

Factor

Comment

Exercise of injected area

Strenuous exercise of a limb within 1 hour of injection will speed insulin absorption.

Clinically significant for regular insulin analogs.

Local massage

Vigorously rubbing or massaging the injection site will speed absorption.

Temperature

Heat can increase absorption rate, including use of a sauna, shower, or hot bath soon after injection.

Cold has the opposite effect.

Site of injection

Insulin is absorbed faster from the abdomen. Less clinically relevant with rapid-acting insulins, insulin glargine, and insulin detemir.

Lipohypertrophy

Injection into hypertrophied areas delays insulin absorption.

Jet injectors and inhaled insulin

Increase absorption rate.

Insulin mixtures

Absorption rates are unpredictable when suspension insulins are not mixed adequately (i.e., they need to be resuspended).

Insulin dose

Larger doses delay insulin action and prolong duration.

Physical status (soluble vs.suspension)

Suspension insulins must be sufficiently resuspended prior to injection to reduce variability.

 

Elimination

 

The kidneys and liver account for the majority of insulin degradation. Normally, the liver degrades 50-60% of insulin released by the pancreas into the portal vein, and the kidneys ~35- 45% (26,29) . When insulin is injected exogenously, the degradation profile is altered since insulin is no longer delivered directly to the portal vein. The kidneys play a greater role in insulin degradation with SQ insulin (~60%), with the liver degrading ~30-40% (30).

 

Because the kidneys are involved in the degradation of insulin, renal dysfunction will reduce the clearance of insulin and prolong its effect. This decreased clearance is seen with both endogenous insulin production (either normalproduction or that stimulated by oral agents) and exogenous insulin administration. Renal function generally needs to be greatly diminished before this becomes clinically significant (31). Clinically, a deterioration in renal function leads to a progressive decline in exogenous insulin requirements and an increased risk of hypoglycemia.

 

PHARMACODYNAMICS

 

The onset, peak, and duration of effect vary among insulin preparations. Insulin pharmacodynamics refers to the metabolic effect of insulin. Commercially available insulins are categorized as rapid-acting, short-acting, intermediate-acting, and long- acting. Insulins currently available in the United States are listed in Table 2. Insulin pharmacodynamics of the various insulins are shown in Table 3. Ranges are listed for the onset, peak and duration, accounting for intra/inter-patient variability. By having patients self-monitor their blood glucose frequently, the patient-specific time-action profile of the specific insulin can be better appreciated. Figures 4-6 show the time-activity profiles for available injectable and inhaled insulins.

 

Table 2. Insulins Commercially Available in the US (Recombinant DNA Origin)

Category/Nameof Insulin

Brand Name (manufacturer)

Preparation(s)

Rapid-Acting

Insulin Lispro

Humalog (Lilly)

Admelog (Sanofi)

Lyumjev (Lilly)

Vial, cartridge, pen

Vial, pen

Vial, pen

Insulin Aspart

Novolog (Novo Nordisk)

Fiasp (Novo Nordisk)

Vial, cartridge, pen

Vial, cartridge, pen

Insulin Glulisine

Apidra (Sanofi-Aventis)

Vial, pen

Technosphere insulin

Afreeza

Inhaler

Short-Acting

Regular Human

Humulin R (Lilly)

Novolin R (Novo Nordisk)

Vial

Vial

Intermediate-Acting

NPH Human

Humulin N (Lilly)

Novolin N (Novo Nordisk)

Vial, pen

Vial, pen

Long-Acting

Insulin Detemir

Levemir (Novo Nordisk)

Vial, pen

Insulin Glargine

Lantus (Sanofi-Aventis)

Basaglar (Lilly)

Toujeo (Sanofi-Aventis)

Vial, cartridge, pen

Pen

Pen

Insulin Glargine-yfgn

Semglee (Viatris)

Vial, pen

Insulin Degludec

Tresiba (Novo Nordisk)

Pen

Insulin Mixtures

NPH/Regular (70%/30%)

Humulin 70/30 (Lilly)

Novolin 70/30 (Novo Nordisk)

Vial, pen

 Vial, pen

Protamine/Lispro (50%/50%)

Protamine/Lispro (75%/25%)

Protamine/Aspart (70%/30%)

Humalog Mix 50/50(Lilly)

Humalog Mix 75/25(Lilly)

Novolog Mix 70/30 (NovoNordisk)

Vial, pen

Vial, pen

Vial, pen

 

Table 3. Insulin Pharmacodynamics  (3–11,28,30,32–35)

Insulin

Onset of action (hr.)

Peak (hr.)

Duration (hr.)

Appearance

Fast-acting Insulin Aspart

16 min

~1

~5

Clear

Insulin Lispro

23-27 min

~ 1-2

~5

Clear

Insulin Lispro-aabc

15-18 minutes

~ 1-2

~4

Clear

Insulin Aspart

21 min

1-3

~5

Clear

Insulin Glulisine

0.25-0.5

0.5-1

~ 4

Clear

Technosphere

within 5 min

15 min

~ 3

Powder

Regular

~ 1

2-4

5-8

Clear

NPH

1-2

4-10

14+

Cloudy

Insulin Detemir

3-4

6-8 (though

relatively flat)

up to 20-24

Clear

Insulin Glargine

1.5

Flat

24

Clear

Insulin Degludec

1

9

42

Clear

Lispro Mix 50/50

0.25-0.5

0.5-3

14-24

Cloudy

Lispro Mix 75/25

0.25-5

0.5-2.5

14-24

Cloudy

Aspart Mix 70/30

0.1-0.2

1-4

18-24

Cloudy

Patient specific onset, peak, duration may vary from times listed in table.

 

Figure 4. Pharmacodynamic Profiles of a Rapid Insulin Analog (insulin lispro) and Regular Insulin (33,36).

Figure 5. Pharmacodynamic Profiles of Faster Aspart and Insulin Aspart (37).

Figure 6. Pharmacodynamic Profiles of Long-Acting and Intermediate-Acting Basal Insulins (38,39).

Dose-Dependent Effect

 

The pharmacodynamics of regular and NPH are particularly affected by the size of the dose (40). Larger doses can cause a delay in the peak and increase the duration of action.

 

INSULIN PREPARATIONS

 

Short-Acting (Prandial or Bolus) Regular Insulin

 

Regular insulin is injected pre-meal to blunt the postprandial rise in glucose levels. It forms hexamers after injection into the SQ space slowing its absorption. Hexameric insulin progressively dissociates into absorbable insulin dimersand monomers. For this reason, regular insulin has a delayed onset of action of 30-60 minutes, and should be injected approximately 30 minutes before the meal to blunt the postprandial rise in blood glucose. Adherence to a 30-minute pre-meal schedule is inconvenient and difficult for many patients.

 

Rapid-Acting (Prandial or Bolus) Insulin Analogs

 

Rapid-acting analogs result from changes to the amino acid structure of human insulin which lead to decreases in hexameric insulin formation after injection into the SQ space. This leads to more rapid dissolution of insulin into monomers, more rapid insulin absorption into the bloodstream, and a shorter duration of action. While on a molar basis rapid-acting insulin analogs have identical in vivo potency compared to regular human insulin, higher peak concentrations are achieved (30). For this reason, when converting from regular to a rapid-acting insulin analog, the dose of insulin may need to be reduced. When compared to regular insulin, the rapid-acting insulin analogs lead to less postprandial hyperglycemia and less late postprandial hypoglycemia (41–43). Injection of rapid-acting insulin analogs 15-20 minutes pre-meal leads to maximal reduction of postprandial glucose excursions (44,45), as compared to 30 or more minutes pre-meal for regular insulin. This shorter interval for insulin injection pre-meal is more convenient for patients and leads to greater adherence with prescribed injection timing guidelines. In patients who areunsure of the amount of carbohydrate to be served for a meal, immediate pre-meal dosing allows more accurate dosing and reduces the risk of hypoglycemia.

 

The use of faster aspart in Medtronic insulin pumps in people with type 1 diabetes was compared to use of insulin aspart in a randomized trial. Faster aspart was non-inferior to aspart with respect to change in A1c from baseline, and no statistically significant difference in rates of severe hypoglycemia was noted between the two arms (46). In a randomized trial of patients on insulin pumps, lispro-aabc was also shown to be non-inferior with respect to change in baseline A1c when compared with insulin lispro, with similar rates of hypoglycemia (47). Both faster aspart and lispro-aabc lower postprandial glucose levels more effectively but cause more frequent infusion site reactions than aspart and lispro, respectively. Based on manufacturer’s recommendations, any rapid-acting insulin analogs, including Fiasp and Lyumjev, can be used in Omnipod pumps (48). For Tandem and Medtronic insulin pumps, only Humalog and Novolog insulin have been approved (49,50).

 

INSULIN LISPRO (HUMALOG)

 

Insulin lispro (Humalog) results from the reversal of the B28 (proline) and B29 (lysine) amino acid sequence of insulin. Insulin lispro has been approved for injection before and immediately after a meal. Post-meal insulin dosing isuseful for parents of young children with type 1 diabetes or for ill, insulin-requiring hospitalized patients, inwhom the amount of carbohydrates consumed at a meal can be unpredictable. When compared with pre-meal regular insulin in prepubertal children, post meal insulin lispro showed no significant differences inpost meal glucose levels, rates of hypoglycemia, or HbA1c (51). In the rare case of severe human insulin allergy, insulin lispro has been shown to be less immunogenic (52). Lispro-aabc includes 2 excipients, citrate and trepostinil that speed insulin absorption by their effects on local blood vessels (34). Citrate increases vascular permeability and treprostinil promotes vasodilation.

 

INSULIN ASPART (NOVOLOG)

 

Insulin aspart differs from human insulin by a substitution of the B28 amino acid proline with aspartic acid. Chemicallyit is B28-aspartic acid-human insulin. Fast-acting aspart adds 2 excipients, niacinamide and L-arginine, to conventional aspart (53). Proposed mechanisms by which niacinamide promotes more rapid insulin aspart absorption include more rapid formation of monomeric insulin, and enhanced local vascular vasodilatation and permeability (54).

 

INSULIN GLULISINE (APIDRA)

 

Insulin glulisine differs from human insulin by changes in the amino acid asparagine at position B3 to lysine and the lysine at position B29 to glutamic acid. Chemically, it is 3B-lysine-29B-glutamic acid-human insulin.

 

INHALED (TECHNOSPHERE) INSULIN (AFREZZA)

 

Inhaled insulin formulations deliver powdered recombinant human regular insulin into the lower airways using an inhaled delivery device. Exubera received FDA approval in 2006 but failed to gain market share and production was discontinued after 1 year. Technosphere insulin (Afrezza) was FDA approved in 2014. Its pulmonary absorption leads to a more rapid absorption than currently available, subcutaneously administered rapid-acting insulin preparations. In subjects with type 2 diabetes, serum insulin levels rise within 5 minutes after inhalation and peak after 17 minutes (55). When compared with pre-meal human regular insulin, technosphere insulin more effectively reduced 4-hour postprandial glucose area under the curve by 52% (56). Only 0.3% of technosphere insulin is detectable in the lungs after 12 hours.

 

Technosphere insulin leads to a dry cough in 19-30% of subjects tested (36,55,57,58). Small reductions in forced expiratory volume (FEV1) are observed in the first 3-6 months of use which are non-progressive for up to 2 years of follow up and reversible after drug discontinuation (42,44,45). The use of technosphere insulin is contraindicated inpatients who smoke or have COPD because of alterations in drug absorption. Spirometry needs to be performed priorto initiation of technosphere insulin, after 6 months, and then annually thereafter, with a 20% or higher decline in FEV1being an indication for drug discontinuation (59).

 

COMPARISONS OF PRANDIAL INSULINS

 

No significant differences in glycemic control have been observed in most studies comparing insulin aspart, insulin lispro, and insulin glulisine. Although insulin glulisine exhibits a more rapid onset of action than either insulin lispro or insulin aspart, this does not translate to meaningful clinical differences between these short-acting analog insulins (60).

 

Faster aspart results in a more rapid onset of action and more glucose lowering within 30 minutes of administration than insulin aspart. However, no significant difference between faster aspart and insulin aspart has been observed in total glucose lowering (61).  Lispro-aabc has a more rapid onset of action and a shorter duration of action compared with lispro (34). Insulin lispro-aabc also has faster absorption than both insulin aspart and faster aspart (62).

 

Intermediate-Acting Insulins (NPH)

 

NPH (Neutral Protamine Hagedorn) insulin, was created in 1936 after it was discovered that the effects of subcutaneously injected insulin could be prolonged by the addition of the protein protamine. NPH insulin is an intermediate-acting insulin, with an onset of action of approximately 2 hours, peak effect 6-14 hours, and duration ofaction 10-16 hours (depending on the size of the dose). Because of its broad peak and long duration of action, NPH can serve as a basal insulin only when dosed at bedtime, or a basal and prandial insulin when dosed in the morning. NPH insulin is available in various combinations with either regular insulin or rapid-acting insulins (Table 2).

 

Long-Acting (Basal) Insulin Analogs

 

Long-acting insulins provide basal insulin coverage. Basal insulins suppress hepatic gluconeogenesis to preventglucose levels from rising during the fasting state in insulin-deficient patients. Among patients with type 1 diabetes, basal insulins additionally prevent ketogenesis.

 

INSULIN GLARGINE (Lantus)

 

Insulin glargine (21A-Gly-30Ba-L-Arg-30Bb-L-Arg-human insulin) contains two modifications to human insulin. Twoarginines are added to the C-terminus of the B chain shifting the isoelectric point of the insulin from a pH of 5.4 to 6.7 (63). This change makes the insulin more soluble at an acidic pH, and insulin glargine is formulated at a pH of 4.0. The second modification is at position A21, where asparagine is replaced by glycine. This substitution prevents deamidation and dimerization that would occur with acid-sensitive asparagine. When insulin glargine is injected into subcutaneous tissue, which is at physiologic pH, the acidic solution is neutralized. Microprecipitates of insulin glargine are formed, from which small amounts of insulin are released throughout a 24- hour period, resulting in a relativelystable level of insulin throughout the day (64). The biological activity of insulin glargine is due to its absorption kinetics and not a different pharmacodynamic activity (e.g., stimulation of peripheral glucose uptake) (65).

 

Insulin glargine should not be mixed in the same syringe with any another insulin or solution because this will alterits pH and thus affect its absorption profile. Glargine has an onset of action of about 2 hours, and a duration ofaction of 20-24 hours. It may be given once daily at any time of day, or twice daily at higher doses (typically more than 50 units daily) to better maintain its relatively flat action profile. Its more consistent rate of absorption andlack of a significant peak action result in reduced nocturnal hypoglycemia when insulin glargine is used atbedtime compared with NPH insulin (66,67).

 

INSULIN DETEMIR (LEVEMIR)

 

Insulin detemir (Levemir) is a long-acting insulin analog in which the B30 amino acid is omitted and a C14 fatty acidchain (myristic acid) is bound to the B29 lysine amino acid. Insulin detemir is slowly absorbed due to its strong association with albumin in the SQ tissue. When it reaches the bloodstream it again binds to albumin delaying its distribution to peripheral tissues. Detemir has an onset of action of about 2 hours, and a duration of action of 16-24 hours. It can be given once or twice daily. Patients who experience a rise in glucose levels in the hours prior to a once daily injection due to the waning action of detemir should use a twice daily dosing regimen.

 

INSULIN DEGLUDEC (TRESIBA)

 

Insulin degludec (Tresiba) is an ultra-long-acting modified human insulin in which the B30 amino acid is omitted and a glutamic acid spacer links a 16-carbon fatty di-acid chain to the B29 amino acid. Deguldec forms multihexamersfollowing SQ injection, leading to a slow release of insulin monomers into the bloodstream and a prolonged duration of action. The half-life of degludec is about 25 hours and its duration of action more than 42 hours. Flat insulin levels are seen within 3 days of the first injection with less daytime variability when compared with glargine insulin (68) . With overall similar HbA1c lowering when compared with glargine insulin, reduced rates of hypoglycemia have been seen with degludec use in type 2 diabetes patients, but not in type 1 diabetes patients (69). No differences in local site reactions, weight gain, or other adverse reactions have been seen with degludec use.

 

In a preapproval cardiovascular outcomes trial of patients with type 2 diabetes with a history of cardiovascular diseaseor at high cardiovascular disease risk, insulin degludec was found to be non- inferior to insulin glargine with respect to the likelihood of major adverse cardiovascular events, including cardiovascular death, nonfatal MI, or nonfatal stroke (70). Insulin degludec was associated with less overall symptomatic hypoglycemia when compared to insulin glargine in a randomized, controlled trial of patients with type 1 diabetes who were at risk for hypoglycemia, with no significant difference in glycemic control between the two treatment arms (71). Similar results were noted in a randomized, controlled trial of patients with type 2 diabetes on insulin and at risk for hypoglycemia (72).

 

COMPARISON OF BASAL INSULINS

 

Compared to NPH, insulin glargine results in significantly less overnight hypoglycemia and a lower rate of hypoglycemic events (73,74). Insulin detemir also results in less overall and nocturnal hypoglycemia compared to NPH (73,75).

 

Differences have also been noted between U-100 insulin glargine and U-300 insulin glargine. In a study of patientswith type 2 diabetes who used mealtime insulin and were on ≥ 42 units insulin daily, U-300 insulin glargine resulted in less nocturnal hypoglycemia compared to U-100 insulin glargine (76). A similar result was seen in a study of patients with type 2 diabetes on basal insulin and oral antihyperglycemic agents (77). In a study of patients with type 2 diabetes who had not previously been treated with insulin, U-300 insulin glargine was associated with a lower risk of hypoglycemia over the study period, although there was no significant difference in the treatment groups in nocturnalhypoglycemia (78). No significant difference in A1C lowering between U-100 glargine and U-300 glargine was noted in these studies (76–78).

 

Pre-mixed Intermediate with Short or Rapid-acting insulins (50/50, 70/30 and 75/25)

 

NPH insulin or protamine added to rapid-acting insulin analogs can be mixed together with regular or rapid-acting insulin analogs in fixed combinations. These insulins thus provide bolus insulin coverage for the meal that follows the injections well as basal coverage from the intermediate-acting component of the insulin. They are given either before a larger breakfast or dinner meal as once daily dosing, or more commonly twice daily before breakfast and dinner. Patients who require basal/bolus insulin replacement but have difficulty with frequently missed insulin dosages maybenefit from a regimen utilizing twice daily mixed insulin. However, given the fixed proportions of mixed insulins and their less physiologic action, there is an increased risk of hypoglycemia using these insulin preparations when compared with basal and pre-meal bolus insulin regimens (79).

 

Follow-on Biologic, and Biosimilar Insulins

 

Relative to the production of other medications, the production of a biologically similar insulin is a more complicated process, which contributes to reduced cost savings in purchasing insulin (68). Basaglar, a “follow-on biologic” insulin of Lantus or insulin glargine, was approved by the FDA in 2015 (80). Similarly, Admelog is a follow-on insulin ofHumalog, or insulin lispro (81). In 2021, Glargine-yfgn (Semglee)became the first biosimilar insulin to be approved in the United States. Biosimilar insulins possess the same biologic and pharmacokinetic properties as the reference insulin. Trials comparing glargine-yfgn to glargine have shown no significant differences in glycemic control or adverse effects, even when the insulins were switched during the study, among participants with type 1 and type 2 diabetes (82–84). Glargine-yfgn has been given the designation of an interchangeable biosimilar insulin, meaning it can be substituted for brand glargine by the pharmacy based on insurance coverage without notification of the prescriber.

 

STORAGE

 

All insulins have an expiration date on the package labeling that applies to insulins that are unopened and refrigerated. Unopened insulin (i.e., not previously used) should be stored in the refrigerator at 36°F- 46°F (2°C- 8°C). Insulin should never be frozen, kept in direct sunlight, or stored in an ambient temperature greater than 86°F (30°C). Exposure to extremes of temperature can lead to loss of insulin effectiveness and a deterioration in glycemic control. Insulin that has been removal from the original vial (i.e., for pump use) should be used within two weeks or discarded. Insulin vials, cartridges, or pens may be kept at room temperature, between 59 °F-86°F (15 °C-30°C), for 28 days, orabout 1 month. Insulin detemir can be stored at room temperature for up to 42 days.

 

Regular insulin, the basal insulin analogs (glargine, detemir, and degludec) and the rapid-acting insulin analogs(lispro, aspart, and glulisine) are clear and colorless and should not be used if they become cloudy or viscous.

 

ADVERSE EFFECTS

 

Hypoglycemia

 

Hypoglycemia is the most serious adverse effect of insulin therapy and the major barrier to achieving glycemic targets in patients with type 1 diabetes and insulin-requiring type 2 diabetes (85). Intensive insulin therapy in patients with type 1 diabetes in the DCCT was associated with a 2-3 fold increase in severe hypoglycemia (SH), defined as hypoglycemia requiring assistance from others (86). In studies of intensive therapy in type 2 diabetes, including the UKPDS, VADT, ADVANCE, and ACCORD trials, intensive therapy resulted in significantly more common SH when compared with standard therapy (29,87–89). SH can cause confusion, motor vehicle accidents, seizures and coma,and is estimated to be a cause of death in 4-10% of patients with type 1 diabetes (90).

 

In one study, the adjusted probability of SH was found to range between 1.02 to 3.04% in patients with type 2 diabetes, depending on clinical complexity and intensity of treatment (91). Patients with type 2 diabetes who have had SH are at increased risk of death regardless of the intensity of their glycemic control. Hypoglycemia increases heartrate, systolic blood pressure, myocardial contractility and cardiac output, which may adversely affect those with diabetes who frequently have underlying coronary artery disease (CAD). Glucose levels below 70 mg/dl have been shown to cause ischemic ECG changes in patients with type 2 diabetes and CAD during continuous glucose and ECG monitoring (92). Hypoglycemia may lead to increased mortality due to the pro-arrhythmic effects of sympathoadrenal activation and hypokalemia (93), or from cardiac repolarization, especially in older patients with underlying cardiac disease.

 

Risk factors for hypoglycemia among insulin-treated patients include older age, longer duration of diabetes, renalinsufficiency, hypoglycemia unawareness, prior hypoglycemia, and lower HbA1c (94–97). Avoidance of hypoglycemia therefore takes on particular importance in older patients, given the greater prevalence of cardiovascular disease, cognitive dysfunction, and higher risk of falls and fractures. To help reduce the incidence of hypoglycemia, the American Diabetes Association (ADA) advises targeting a higher HbA1c of less than 8% in patients who are older, with a longer duration of disease, more comorbidities, frequent hypoglycemia, and underlying cardiovascular disease (98). All patients receiving insulin should learn to recognize the symptoms of hypoglycemia and how best to treat low glucose levels.

 

The use of continuous glucose monitoring (CGM) in adolescent, young and older adults with type 1 diabetes has been shown to reduce the frequency of hypoglycemia, while lowering hemoglobin A1c (99,100). Whether CGM use reduces death from hypoglycemia remains to be determined.

 

Weight Gain

 

Weight gain is a common side effect of insulin therapy. In part, the weight gain can be a result of frequenthypoglycemic episodes in which patients consume extra calories to treat the low glucose level and oftenovereat in response to hunger. Additionally, amelioration of glycosuria can prevent the loss of calories in the urine. One of the anabolic effects of insulin is to promote the uptake of fatty acids into adipose tissue. The amount of weight gain in the DCCT (type 1 patients) and UKPDS (type 2 patients) associated with insulin therapy was 4.6 kg and 4.0 kg respectively (86,87). Less weight gain is encountered with detemir insulin than withNPH or glargine insulin (101,102). The etiology of lower weight gain with detemir when compared with NPH or glargine is not entirely understood (103). Basal insulin added to oral antihyperglycemic agents leads to less weightgain than either biphasic insulin aspart or prandial aspart insulin (79). Lispro mix 75/25 insulin leads to greaterweight gain than glargine insulin when added to oral antihyperglycemic agents (104).

 

Local Reactions

 

True allergic reactions and cutaneous reactions are rare with human insulin and insulin analogs. Hypersensitivity reactions rarely develop in response to the insulin or one of its additives (protamine for example) and can result in local erythema, pruritus, a wheal or more systemic reactions including anaphylaxis. Successful approaches to insulin allergies include continuous subcutaneous insulin infusions, and use of lispro insulin which appears to be less allergenic (52,105). Lipoatrophy was common with use of less pure and animal insulins, but is now rarely seen with insulin analogues and believed to be immune-mediated. Cases of lipoatrophy have been reported with the use of glargine, aspart, and lispro insulins (106). To avoid the lipohypertrophic effects of insulin, patients should be instructed to rotate their insulin injection sites, preferably rotating within one area and not reusing for one week.

 

Mitogenic Properties

 

Several retrospective, observational studies have shown correlations between insulin dose and cancer risk for most insulin types (human insulin, aspart, lispro or glargine) (107–109). These observational studies assessed large patient databases and have significant, inherent limitations, such as the potential for different pre-treatment characteristics of the groups, selection bias, the small numbers of cancer cases found, and short duration of follow-up. Meta-analyses of studies comparing exogenous insulin to non-insulin antihyperglycemic therapies have shown associations of insulin with several cancers (15,16). However, there are also inherent limitations to such analyses. In a randomized, 5-year, open-label trial comparing the progression of retinopathy in NPH and insulin glargine users, no increased risk ofcancer was found in the 1,017 patient sample (102). In an analysis of 31 randomized controlled trials from the Sanofi-Aventis safety database (phase 2, 3, and 4 studies), insulin glargine was not associated with an increased risk of cancer (110). The 7-year, randomized ORIGIN trial assessed the cardiovascular effects of insulin glargine versus standard care in more than 12,500 individuals with diabetes or pre-diabetes and found no increased risk of all- cancer-combined or of cancer mortality among glargine-treated individuals (111). A review of large epidemiologicstudies did not find evidence of an increased risk of malignancy among glargine- treated patients when compared with other insulin therapies (16).

 

Cardiovascular Disease

 

Among intensively controlled patients in the VADT (Veterans Affairs Diabetes Trial), ADVANCE (Action in Diabetes and Vascular Disease: Preterax and Diamicron MR Controlled Evaluation), and ACCORD (Action to ControlCardiovascular Risk in Diabetes), a higher proportion (41–90%) required insulin therapy than among the standard control groups (24–74%) (29,88,89). Nonsignificant reductions in cardiovascular events were seen with intensive diabetes control when compared with standard control in ADVANCE, ACCORD, and VADT respectively. An increased mortality rate was observed in ACCORD after 3.5 years of intensive therapy when patients were targeted to an HbA1c of less than 6.0%, 73% of whom received insulin. However, mortality was not temporally associated with severe hypoglycemia. The results of ACCORD support less aggressive diabetes management among patients at high risk for a cardiovascular event. The 7-year, randomized ORIGIN trial assessed the cardiovascular effects of insulin glargine versus standard care in more than 12,500 individuals with diabetes or pre-diabetes and found no increasedrisk of cardiovascular events or of cardiovascular mortality among glargine-treated individuals (111). In a retrospective study of more than 57,000 insulin-treated persons with type 2 diabetes followed for 4 years, treatment with combined long and short-acting insulins compared to long-acting insulin alone was associated with increased all-cause mortality, but fewer myocardial infarctions, and no difference in major cardiovascular events or cardiovascular mortality (112).

 

TYPES OF REGIMENS

 

General Principles

 

TYPE 1 DIABETES

 

Autoimmune beta cell destruction results in a progressive decline in insulin production in patients with type 1 diabetes who eventually require insulin for survival. Patients with type 1 diabetes generally require a replacementdose of 0.5-1.0 units per kg of body weight per day of insulin (113).

 

During the early stages of type 1 diabetes, patients commonly require less insulin because remaining beta cells still produce some insulin; during their “honeymoon period” insulin requirements can be in the range of 0.2-0.6 units per kg per day (113,114). Intensive insulin therapy (defined as 3 or more insulin injections daily or insulin pump therapy) is indicated for patients with type 1 diabetes to provide better glycemic control with less glucose variability than 1 or 2 daily injections, and reduce the development and progression of microvascular and macrovascular complications (86,115,116).

 

TYPE 2 DIABETES

 

The slowly progressive beta cell loss in patients with type 2 diabetes means many patients with type 2 diabetes will eventually require insulin therapy to attain adequate glycemic control. Initiation of basal insulin can be consideredin any of the following situations: 1) a patient with an HbA1C > 11% with concern for insulin deficiency; 2)a patient without atherosclerotic cardiovascular disease and/or chronic kidney disease with an HbA1C above goal after 3 months of metformin monotherapy; 3) a patient with atherosclerotic cardiovascular disease and/or chronic kidney disease with an HbA1C above goal after 3 months of treatment withmetformin, a GLP-1 receptor agonist, and/or an SGLT2 inhibitor (98). Since type 2 diabetes is associated with insulin resistance, insulin requirements can exceed 1 unit/kg/day. When initiating insulin therapy in patients with type 2 diabetes, basal insulin is often used in combination with other non-insulin antihyperglycemic medications a patient is taking. An intermediate or long-acting insulin (e.g., NPH, glargine, detemir, or degludec) is added at bedtime, and the dose titrated to attain a target fasting glucose (117).

 

Basal insulin is effective at lowering HbA1c when added to oral hypoglycemic agents starting at a dose of 10 U daily or 0.2 U/kg. When used in patients uncontrolled on oral agents, basal insulin lowers HbA1c 1.2–1.5% (79,102). In these treat-to-target studies, patients were instructed to titrate their basal insulin dosages up every 2–3 days by 1–4 units based on algorithms to achieve fasting plasma glucose (FPG) levels in the 70- to 126mg/dl range. Those studies targeting a FPG < 108 mg/dl achieve modestly better success in achieving an HbA1c < 7% (63.2 vs. 52%) than those targeting a FPG of less than 126 mg/dl, with mildly higher rates of hypoglycemia (79,102).

 

Basal insulin, by suppressing hepatic glucose output during the night, will control the fasting blood glucose (FPG), while concomitant use of other antihyperglycemic medications control postprandial glucose levels throughout the day (118). A starting dose of 10 units of basal insulin is commonly utilized, though starting a dose of 0.1-0.2 units/kg will more rapidly attain the target fasting glucose level (119). In patients whose fasting glucose levels becomewell controlled with basal insulin, but whose glucose levels rise significantly higher later in the day with apersistently elevated HbA1C, prandial insulin is indicated. At this point, the patient is experiencing beta- cell failure. If the patient is taking an insulin secretagogue (e.g., a sulfonylurea or meglitinide) it should be discontinued, as insulin will now be replaced exogenously. However, other agents not having a predominantly insulin-stimulating effect should be continued to address insulin resistance and reduce insulin requirements.

 

GOALS OF THERAPY

 

Before starting a patient on insulin, or adjusting their current insulin therapy, it is important to establish glycemic goals tailored to the patient. The American Diabetes Association currently recommends individualized glycemic goals (98).Those with a longer duration of diabetes, shorter life expectancy, presence of important comorbidities orestablished vascular complications, and at higher risk of hypoglycemia should have higher glycemic targets, with an A1C of < 8% reasonable for those with the least to gain from more intensive control and at highestrisk for adverse outcomes from hypoglycemia. For the majority of patients who are otherwise healthy, glycemic targets include the following: preprandial plasma glucose 80-130 mg/dl; postprandial plasma glucose <180 mg/dl; and A1C <7% (98).

 

In the DCCT, retinopathy initially worsened during the first year in patients with type 1 diabetes who received intensive therapy (86). This was associated with rapid lowering of glucose levels. Thus, in patients with proliferative retinopathy or those with underlying non-proliferative diabetic retinopathy and a high A1C (e.g., >10%), slower lowering of glucose is warranted. Another example of individualizing glycemic goals is a patient with hypoglycemic unawareness, in whom glycemic goals should be less aggressive to reduce the frequency of severe hypoglycemia  (115).

 

REPLACEMENT STRATEGIES

 

Physiologic Insulin Replacement

 

A functioning pancreas releases insulin continuously, to supply a basal amount to suppress hepatic glucose outputand prevent ketogenesis between meals and overnight, and also releases a bolus of insulin prandially to promote glucose utilization after eating (108) . Replacing insulin in a manner that attempts to mimic physiologic insulin release is commonly referred to as basal/bolus insulin therapy. Physiologic replacement requires multiple daily injections (3 or more) or the use of an insulin pump. Basal insulin requirements are approximately 40-50% of the total daily amount. Prandial insulin is 50-60% of the total daily insulin requirement administered before meals (114) . Providing basal-bolus insulin regimens allows patients to have flexibility in their mealtimes and achieve better glycemic control.

 

Non-Physiologic Insulin Replacement

 

When insulin is given once or twice daily, insulin levels do not mimic physiologic insulin release patterns. For people with type 2 diabetes, in whom bolus insulin replacement is not as critical, once or twice daily basal insulin injection regimens often work well with reasonable glycemic control achieved when combined with non-insulin agents that control postprandial glucose levels.

 

In patients with type 2 diabetes, a starting daily basal insulin dose can be calculated by multiplying 0.1 or 0.2 by thepatient’s weight in kilograms and increased after taking into account factors such as the severity of hyperglycemia (98). The basal insulin dose in type 2 patients is adjusted to attain a target fasting glucose level. The patient’s other non-insulin anti-hyperglycemic agents then can better control daytime glucose levels.

 

The current American Diabetes Association guidelines recommend that if a patient’s A1C is not at goal despite the use of basal insulin attaining target fasting glucose levels, then an additional injection of prandial insulin, with astarting dose comprised of 4 units or 10% of the daily basal insulin dose, can be added before the biggest meal of the day. Prandial insulin can be titrated based on blood glucose measurements by 10-15% twice a week. If glycemic control is suboptimal, then more prandial insulin injections can be added before other meals (98).

 

EXAMPLES OF REGIMENS

 

Once Daily Insulin Regimen (for patients with type 2 diabetes on oral agents)

 

NPH (Figure 7), insulin glargine (Figure 8), or insulin detemir are most often given at bedtime. However, given their longer duration of action, insulin glargine and insulin degludec can be administered anytime of the day (101). For patients who eat large amounts of carbohydrates at dinner, an insulin mixture, regular and NPH or a premixed insulin, can be given prior to dinner (Figure 9).

 

Figure 7. PM NPH Administration.

Figure 8. Glargine Administration.

Figure 9. NPH and Regular Insulin at Dinner.

Twice-Daily Insulin Regimen (Split-Mixed and Pre-Mixed Regimens)

 

Two-thirds of the insulin dose is typically given in the morning before breakfast and one-third is given before dinner. Premixed insulins can be used or a mixture of a short-acting insulin (e.g., regular, insulin aspart/glulisine/lispro) and an intermediate-acting insulin (e.g., NPH) (Figure 10) (114) .

 

Figure 10. NPH and Twice a Day Regular Insulin.

 

2/3 total daily dose at breakfast: given as 2/3 NPH and 1/3 Regular (or insulin aspart/glulisine/lispro) 1/3 total daily dose at dinner: divided in equal amounts of NPH and Regular (or insulin aspart/glulisine/lispro)

 

For patients who experience nocturnal hypoglycemia when NPH is administered at dinner with a short-acting insulin, moving the NPH dose to bedtime helps reduce the risk for nocturnal hypoglycemia (120). Conversely, NPH at dinner can result in fasting hyperglycemia due to dissipation of insulin activity and the early morning rise in counter-regulatory hormones cortisol and growth hormone (the dawn phenomenon). Moving the NPH dose to bedtime can also help resolve this problem (121)  (Figure 11). An obvious limitation to using premixed insulin is reduced flexibility in dosing; if the dose is adjusted, both types of insulin in the mixture will be adjusted.

 

Figure 11. Twice a Day NPH and Regular.

Multiple Daily Insulin Injection Regimen: Basal plus Prandial Insulin

 

Many different types of regimens are possible with multiple daily injections. Regular, insulin aspart, glulisine and lispro are used to provide prandial insulin. NPH, insulin glargine, insulin detemir, and insulin degludec are used to provide basal insulin.

 

Regular, insulin aspart/glulisine/lispro before meals and NPH, insulin glargine, insulin detemir, or insulin degludec at bedtime (hs) (Figure 12, 13).

 

Insulin aspart/glulisine/lispro before meals and NPH twice daily (breakfast and bedtime) (Figure 14).

 

Figure 12. Bedtime NPH and Regular Insulin with Meals.

Figure 13. Bedtime Glargine Insulin and Lispro/Aspart with Meals.

Figure 14. NPH Twice a Day and Lispro/Aspart with Meals.

 

Insulin Pump Therapy

 

Insulin pump or continuous subcutaneous insulin infusion (CSII) therapy is another option for intensive insulin therapy using only rapid-acting insulin. Insulin pump therapy is indicated in patients with type 1 diabetes, and in thosewith markedly insulin-deficient type 2 diabetes (122). Patients initiated on insulin pump therapy need to have been trained in the components of intensive diabetes management or will not gain significant benefit from conversion to insulin pump therapy. The components of intensive diabetes management include knowledge of carbohydrate counting and adjustments in the insulin bolus dose based on the carbohydrate content of meals and snacks, the measured glucose level, and the amount and duration of exercise. Some insulin pumps are able to deliver insulinboluses in as low as 0.01-unit increments, ideal for patients who are insulin sensitive. The basal insulin infusion can be delivered in as low as 0.001-unit increments and can be adjusted based on an individual patient’s needs. Basal rate requirements are typically higher in the early morning hours to counter the dawn rise in glucose levels and lower in the afternoon when patients are more active and overnight when patients are at rest. Temporary basal rates can be programmed to be increased during times of inactivity or illness when insulin requirements are higher, and decreased when physically active and insulin requirements are reduced.

 

The bolus calculator function of insulin pumps helps patients determine insulin bolus doses required for the carbohydrate content of foods and the measured glucose level. After a patient enters this information into the pump, a recommended bolus dose is displayed by the pump based on the patient’s insulin-to-carbohydrate ratio and insulin sensitivity factor. This function is especially helpful when a patient needs to determine the amount of supplementalinsulin required to correct a high postprandial glucose level. The pump takes into account the active insulin remaining from the pre-meal bolus (insulin on board), and recommends a reduced corrective supplemental insulin dose, thereby preventing insulin stacking and hypoglycemia.

 

Potential benefits of insulin pumps include less weight gain, less hypoglycemia, and lowering of hemoglobin A1c levels when compared to multiple daily injections (123–126). The addition of continuous glucose monitoring to patients on insulin pumps has been shown to further improve glycemic control and reduce the frequency of symptomatic and severe hypoglycemia. Insulin pumps are available with a threshold suspend function which can discontinue the basal insulin infusion for up to a period of 2 hours when the monitor detects a low glucose level that is untreated. This prevents a further decline in glucose levels (127).

 

To date, several hybrid closed loop (HCL) systems exist. The Medtronic 670G and 770G HCL systems have an “Auto Mode,” in which the basal rate of the insulin pump is adjusted up or down every 5 minutes based on data from the continuous glucose monitoring (CGM) system to achieve a target glucose of 120 mg/dl (128). For the Tandem X2 HCL system, not only is the basal rate of the pump suspended, increased, or decreased every 5 minutes based on CGM data, but automatic bolus doses of insulin, which are comprised of 60% of a calculated corrective bolus dose, are administered up to every hour, to target a glucose of 110 mg/dl (129).  Finally, in the Omnipod 5 HCL system, different target blood glucose values can be set, and basal rates are automatically adjusted (130). It should be noted that once in the automated insulin delivery mode, for Medtronic HCL systems and the Omnipod 5 system, adjustments to manual basal rates do not affect the amount of insulin delivered during basal insulin delivery. For the Medtronic HCL systems, the insulin to carbohydrate ratio and active insulin time can be adjusted, and a temporary target glucose of 150 mg/dl can be selected to better prevent hypoglycemia as during exercise or when fasting for a medical or surgical procedure. For the Omnipod 5, the insulin to carbohydrate ratio, correction factor, and active insulin time can be adjusted. For the Tandem X2 HCL system, active insulin time and target blood glucose cannot be modified, but the basal rate, insulin to carbohydrate ratio, and correction factor can be adjusted (130,131).

 

Timing of Prandial Insulin Injections

 

The onset of action of regular insulin is approximately 30 minutes; while insulin aspart/glulisine/lispro begin to lower glucose levels within about 15 minutes after a subcutaneous bolus is given. Ultra-rapid-acting Lispro-aabc and Faster aspart can be measured in the blood within 1-2.5 minutes after subcutaneous injection. The timing of the pre-meal insulin bolus can be reduced when the measured glucose level is low and lengthened when hyperglycemia is present before eating. To best match the insulin action with the glycemic effect of meals, regular insulin is optimally given 30 minutes before the meal, the rapid-acting insulins 15-20 minutes before the meal, and the ultra-rapid-acting insulins 0-2 minutes before meals. When dosed immediately before a solid mixed meal tolerance test, lispro-aabc resulted in a lower postprandial glucose compared with insulin lispro, over 2 and 5 hours (34). Insulin pumps and multi-dose insulin injection regimens using basal analog insulin combined with a rapid-acting or ultra-rapid-acting insulin provide patients with the greatest flexibility of varying the time of meals without sacrificing an increased risk of hypoglycemia, when compared with NPH-based insulin regimens.

 

ADJUSTMENTS

 

Insulin doses should be adjusted to achieve glycemic targets. Typically, a 10-20% increase or decrease in an insulin dose is appropriate, based on the degree of hyper- or hypoglycemia, and the insulin sensitivity of the patient.Hypoglycemia that is frequent or severe should prompt an immediate reduction in the responsible insulin dose. Increases to insulin doses should be based on the occurrence of consistently elevated glucose levels at a particulartime of day, rather than periodic glucose elevations that are more likely diet-mediated.

 

Adjustment of Intermediate to Long-Acting Insulin

 

When a dose of intermediate or long-acting insulin is adjusted, it is recommended to wait at least 3-5 days before further changes in the dose to assess the response (114) .

 

Adjustment of Once-Daily Evening Insulin

 

Basal insulin can be started either using 10 units or 0.1-0.2 units/kg body weight at bedtime. The FPG is used toadjust the intermediate (NPH), long-acting insulin (glargine, detemir, or degludec) given in the evening.Algorithms provided to patients to adjust their basal insulin dose based on fasting glucose levels have beenshown to improve glycemic control (132). The algorithm should target the fasting glucose range of 80-130mg/dl (98). An example of a forced titration schedule is show below (Table 4):

 

Table 4. Forced Titration Algorithm

Fasting Glucose the past 3 Days

Increase in Basal Insulin (units)

80-130

0

130-159

2

160-189

4

190-220

6

Over 220

8

Decrease dose by 2-4 units for any glucose level < 80

 

Lower dose adjustments are used for more insulin sensitive patients (usually type 1 patients) and higher doses for more insulin resistant patients (usually those with type 2 diabetes). A simple algorithm for patients with type 2diabetes recommends adjusting the basal insulin dose by 2 units every 2 to 3 days if fasting glucose levelsare consistently above the target upper range (98).

 

Supplemental Insulin for Correction of Hyperglycemia

 

Regular insulin, the rapid-acting insulins aspart/glulisine/lispro or the ultra-rapid-acting insulins Lispro-aabc and Faster aspart can be used to correct high glucose levels (133). In type 2 patients, 1-2 units of insulin will lower the blood glucose by 30-50 mg/dl. A commonly used correction insulin regimen which targets a glucose of 100 mg/dlpre-meal and 150 mg/dl at bedtime is shown below (Table 5).

 

Table 5. Basal insulin Forced Titration Algorithm

Time

Breakfast

Lunch

Dinner

Bedtime

Blood Glucose

Extra units of Short or Rapid-

acting insulin

Extra units of Short or Rapid-

acting insulin

Extra units of Short or Rapid-

acting insulin

Extra units of Short or Rapid-

acting insulin

80-150

0

0

0

0

151-200

2

2

2

0

201-250

4

4

4

2

251-300

6

6

6

4

301-350

8

8

8

6

351-400

10

10

10

8

Over 400

12

12

12

10

 

The rule of 1800 can be used to approximate the amount that 1 unit of supplemental insulin will lower the glucose, alsotermed the insulin sensitivity factor (ISF), using the total daily dose (TDD) of insulin:

 

Calculation of the insulin sensitivity factor (ISF):

ISF= 1800/TDD

 

An individual using 60 units of insulin each day would have a calculated ISF of 1:30, and would use 1 unit ofsupplemental insulin for every 30 mg/dl the glucose is above the glucose target (usually 100 mg/dl before meals) as a starting supplemental dose (123).

 

For example, if this person’s pre-meal glucose was 280 mg/dl, 6 units of supplemental insulin would be added to their usual dose of pre-meal insulin to decrease glucose by 180mg/dl.

 

Carbohydrate Counting

 

In patients on set dose of prandial insulin, post meal glucose variability can be controlled by having patients keep the carbohydrate content of the meal similar at mealtimes from day to day. A more sophisticated type of prandial insulinregimen is one in which a patient doses their prandial insulin based on the number of carbohydrates eaten at the meal. By learning how to count carbohydrates, and dosing their insulin accordingly, patients are afforded flexibility in the carbohydrate content of their meals. Adjusting the prandial insulin dose based on the accurately-assessed carbohydrate content of the meal will reduce glucose. The rule of 500 can be used to approximate the amount of carbohydrates covered by 1 unit of prandial insulin, termed the insulin to carbohydrate ratio (ICR), using the total daily dose (TDD) of insulin:

 

Calculation of the insulin to carbohydrate ratio:

ICR= 500/TDD

 

For example, for a patient using 60 units of insulin per day, the ICR would be 500/60 or approximately 1:8, or 1 unit for each 8 grams of carbohydrate in the meal or snack.

 

This ratio is adjusted based on post meal glucose levels and may be different for each meal. The ICR is adjusted toattain post meal glucose levels in the target range (usually 100-180 mg/dl). The ICR can also be used forsnacks (134) . Carbohydrate counting can be challenging for some patients. Education in medical nutrition therapy is critical for patients on insulin.

 

A comprehensive diabetes education class that teaches self-management skills, such as how to dose prandial insulin by matching it to the amount of carbohydrate intake is an excellent resource to facilitate patients in adopting an intensive insulin therapy regimen (135).

 

Adjustments for Exercise

 

Exercise improves insulin sensitivity. Thus, when a patient exercises, it is often necessary to decrease insulin delivery (and/or increase caloric intake) to prevent hypoglycemia. For morning exercise, the pre-breakfast insulin dose should be reduced (by about 25%) depending on the duration and intensity of the exercise. For late-morning/early-afternoon and evening exercise, the pre-lunch and pre-dinner insulin dose should be reduced respectively (136). A more recent consensus statement notes that nutritional insulin should be decreased between 25-75%, depending on the intensity of exercise planned after the meal, with a 25% nutritional insulin dose reduction prior to low-intensity exercise, and a 75% nutritional insulin dose reduction prior to high-intensity exercise (137). In addition, for individuals with type 1 diabetes, before exercise is undertaken, different glycemic thresholds can be set at which point carbohydrates should be ingested, depending on risk of hypoglycemia and intensity of exercise, with a higher glucose goal for the more prolonged exercise and/or for those at highest risk of hypoglycemia (138). The effect of exercise on insulin sensitivity can last for many hours; so more than 1 insulin dose may need to be adjusted. After more prolonged exercise, the bedtime long-acting insulin dose may need to be reduced should such exercise lead to a pronounced fall in overnight glucose levels.  In patients on insulin pumps, temporary basal rate reductions can be employed starting 0-60 minutes before exercise to prevent exercise-induced hypoglycemia. The Tandem X2 and OmniPod 5 automated insulin delivery systems have an “exercise/activity” modes, that target a higher glucose goal within the glucose range of the automated delivery system. The Medtronic automated insulin delivery system has a temporary target glucose level of 150 mg/dl, that can be used during exercise.

 

SELF-MONITORING OF BLOOD GLUCOSE

 

Self-monitoring of blood glucose (SMBG) allows patients and physicians to recognize glucose trends to guideinsulin dosage adjustments. In those using short or rapid-acting inulin, SMBG also provides a patientwith the information needed to give an accurate supplemental insulin dose to return an elevated glucose level back to the target glucose range. Studies in patients with type 1 diabetes have shown aprogressive reduction in hemoglobin A1C levels with more frequent glucose monitoring (139) Currently, theADA recommends that patients with diabetes on multiple daily injections of insulin or on an insulin pump check blood glucose before eating, exercise, and bedtime, for symptoms of hypoglycemia, and periodically after meals. For patients with type 2 diabetes not on multiple daily injections of insulin, no specific frequency of SMBG isrecommended but rather it is recommended that SMBG and its assessment be a part of patients’ treatmentand management plan (139).

 

Most glucose meters are now plasma-referenced, correlating better to the ADA’s glycemic goals. Plasma glucose concentrations are approximately 10-15% higher than whole blood glucose concentrations (140) .

 

CGM, which measures interstitial glucose, is available in 2 forms: an intermittent or “flash” CGM system and real-timeCGM systems (141). To date, the intermittent CGM system and one of the real-time CGM systems do not require calibration with blood glucose. Intermittent CGM has been associated with less time spent in hypoglycemia in patients with type 1 diabetes and in patients with type 2 diabetes (142,143) , and the real-time CGM systems have beenassociated with improved glycemic control, more so when used consistently, and less time spent inhypoglycemia, and less severe hypoglycemia in patients with type 1 diabetes (144,145).

 

SICK DAY GUIDELINES

 

A common misconception among patients is that if they are sick enough that they do not eat or they vomit, they should not take their anti-hyperglycemic medications, insulin included. Patients who are ill should be instructed to continue their basal insulin therapy, maintain fluid intake, eat smaller meals as tolerated, and test their glucoselevels every 1-4 hours (ketones as well for people with type 1 diabetes when glucose levels are over 200 mg/dl).Supplemental insulin doses to correct hyperglycemia can be given up to every 4 hours as needed for persistent hyperglycemia, or more often when the insulin on board from an insulin pump or a smart insulin pen is taken into account. For patients using the bolus calculator function of their insulin pump, the recommended bolusdose to correct an elevated glucose level automatically takes into account the insulin on board from priorinsulin boluses. If the glucose is >240 mg/dl with large ketonuria, patients should contact their provider immediately,or proceed to an emergency room for treatment of ketoacidosis using intravenous fluids and insulin. Sick day guidelines can be found online (146).

 

ACKNOWLEDGEMENTS

 

The prior version of this chapter was extensively modified based on a previous chapter written by Lisa Kroon, PharmD, CDE, Ira D. Goldfine, M.D. and Sinan Tanyolac, M.D.

 

REFERENCES

 

  1. Lawrence M. Landmarks in Insulin Research. Front Endocrinol [Internet]. 2011 [cited 2022 Nov 25];2. Available from: https://www.frontiersin.org/articles/10.3389/fendo.2011.00076
  2. Richter B, Neises G. “Human” insulin versus animal insulin in people with diabetes mellitus. Cochrane Database Syst Rev. 2005 Jan 24;2005(1):CD003816.
  3. FDA. 020563s115lbl.pdf [Internet]. Humalog. [cited 2022 Nov 25]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2013/020563s115lbl.pdf
  4. FDA. 021017s072lbl.pdf [Internet]. Humalog 75/25. [cited 2022 Nov 25]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2013/021017s072lbl.pdf
  5. FDA. 203314lbl.pdf [Internet]. Tresiba. [cited 2022 Nov 25]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2015/203314lbl.pdf
  6. FDA. 021172s034lbl.pdf [Internet]. NovoLog Mix 70/30. [cited 2022 Nov 25]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2007/021172s034lbl.pdf
  7. FDA. 021536s037lbl.pdf [Internet]. Levemir. [cited 2022 Nov 25]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2012/021536s037lbl.pdf
  8. FDA. 021081s072lbl.pdf [Internet]. Lantus. [cited 2022 Nov 25]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/021081s072lbl.pdf
  9. FDA. 021629s015lbl.pdf [Internet]. Apidra. [cited 2022 Nov 25]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2008/021629s015lbl.pdf
  10. FDA. 020986s082lbl.pdf [Internet]. Novolog. [cited 2022 Nov 25]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2015/020986s082lbl.pdf
  11. FDA. 21018lbl.pdf [Internet]. Humalog Mix 50/50. [cited 2022 Nov 25]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/1999/21018lbl.pdf
  12. Correlations of receptor binding and metabolic and mitogenic potencies of insulin analogs designed for clinical use. | Diabetes | American Diabetes Association [Internet]. [cited 2022 Nov 25]. Available from: https://diabetesjournals.org/diabetes/article/49/6/999/11547/Correlations-of-receptor-binding-and-metabolic-and
  13. Najjar SM, Perdomo G. Hepatic Insulin Clearance: Mechanism and Physiology. Physiology. 2019 May 1;34(3):198–215.
  14. Müssig K, Staiger H, Kantartzis K, Fritsche A, Kanz L, Häring HU. Type 2 diabetes mellitus and risk of malignancy: is there a strategy to identify a subphenotype of patients with increased susceptibility to endogenous and exogenous hyperinsulinism? Diabet Med. 2011;28(3):276–86.
  15. Karlstad Ø, Starup-Linde J, Vestergaard P, Hjellvik V, T. Bazelier M, K. Schmidt M, et al. Use of Insulin and Insulin Analogs and Risk of Cancer — Systematic Review and Meta-Analysis of Observational Studies. Curr Drug Saf. 2014 Jan;8(1):333–48.
  16. Janghorbani M, Dehghani M, Salehi-Marzijarani M. Systematic Review and Meta-analysis of Insulin Therapy and Risk of Cancer. Horm Cancer. 2012 Aug 1;3(4):137–46.
  17. Wu JW, Filion KB, Azoulay L, Doll MK, Suissa S. Effect of Long-Acting Insulin Analogs on the Risk of Cancer: A Systematic Review of Observational Studies. Diabetes Care. 2016 Jan 6;39(3):486–94.
  18. Insulin Degludec Is a New Generation Ultra-Long Acting Basal Insulin Designed To Maintain Full Metabolic Effect While Minimizing Mitogenic Potential | American Diabetes Association [Internet]. [cited 2022 Nov 27]. Available from: https://professional.diabetes.org/abstract/insulin-degludec-new-generation-ultra-long-acting-basal-insulin-designed-maintain-full
  19. Schernthaner G. Immunogenicity and Allergenic Potential of Animal and Human Insulins. Diabetes Care. 1993 Dec 1;16(Supplement_3):155–65.
  20. Vardar B, Kızılcı S. Incidence of lipohypertrophy in diabetic patients and a study of influencing factors. Diabetes Res Clin Pract. 2007 Aug 1;77(2):231–6.
  21. Insulin Basics | ADA [Internet]. [cited 2022 Nov 27]. Available from: https://diabetes.org/healthy-living/medication-treatments/insulin-other-injectables/insulin-basics
  22. Pearson SM, Trujillo JM. Conversion from insulin glargine U-100 to insulin glargine U-300 or insulin degludec and the impact on dosage requirements. Ther Adv Endocrinol Metab. 2018 Apr 1;9(4):113–21.
  23. Pettis RJ, Muchmore D, Heinemann L. Subcutaneous Insulin Administration: Sufficient Progress or Ongoing Need? J Diabetes Sci Technol. 2019 Jan 1;13(1):3–7.
  24. Variability of Insulin Absorption and Insulin Action [Internet]. [cited 2022 Dec 17]. Available from: https://www.liebertpub.com/doi/epdf/10.1089/152091502320798312
  25. Gradel AKJ, Porsgaard T, Lykkesfeldt J, Seested T, Gram-Nielsen S, Kristensen NR, et al. Factors Affecting the Absorption of Subcutaneously Administered Insulin: Effect on Variability. J Diabetes Res. 2018 Jul 4;2018:1205121.
  26. Braak EWT, Woodworth JR, Bianchi R, Cerimele B, Erkelens DW, Thijssen JHH, et al. Injection Site Effects on the Pharmacokinetics and Glucodynamics of Insulin Lispro and Regular Insulin. Diabetes Care. 1996 Dec 1;19(12):1437–40.
  27. Nosek L, Coester HV, Roepstorff C, Thomsen HF, Kristensen NR, Haahr H, et al. Glucose-Lowering Effect of Insulin Degludec is Independent of Subcutaneous Injection Region. Clin Drug Investig. 2014 Sep 1;34(9):673–9.
  28. Home PD, Ashwell SG. An overview of insulin glargine. Diabetes Metab Res Rev. 2002;18(S3):S57–63.
  29. Duckworth W, Abraira C, Moritz T, Reda D, Emanuele N, Reaven PD, et al. Glucose Control and Vascular Complications in Veterans with Type 2 Diabetes. N Engl J Med. 2009 Jan 8;360(2):129–39.
  30. Kennedy MSN, Masharani U. Pancreatic Hormones & Antidiabetic Drugs. In: Katzung BG, editor. Basic & Clinical Pharmacology [Internet]. 14th ed. New York, NY: McGraw-Hill Education; 2017 [cited 2022 Dec 18]. Available from: accessmedicine.mhmedical.com/content.aspx?aid=1148439639
  31. Rabkin R, Ryan MP, Duckworth WC. The renal metabolism of insulin. Diabetologia. 1984 Sep;27(3):351–7.
  32. Koda-Kimble MAnne. Applied therapeutics : the clinical use of drugs [Internet]. 9th ed. Philadelphia: Wolters Kluwer Health/Lippincott Williams & Wilkins; 2009. Available from: http://catdir.loc.gov/catdir/enhancements/fy0912/2008024503-t.html
  33. Roach P, Woodworth JR. Clinical Pharmacokinetics and Pharmacodynamics of Insulin Lispro Mixtures. Clin Pharmacokinet. 2002 Nov 1;41(13):1043–57.
  34. Leohr J, Dellva MA, LaBell E, Coutant DE, Klein O, Plum-Moerschel L, et al. Pharmacokinetic and Glucodynamic Responses of Ultra Rapid Lispro vs Lispro Across a Clinically Relevant Range of Subcutaneous Doses in Healthy Subjects. Clin Ther. 2020 Sep 1;42(9):1762-1777.e4.
  35. Heise T, Hövelmann U, Brøndsted L, Adrian CL, Nosek L, Haahr H. Faster-acting insulin aspart: earlier onset of appearance and greater early pharmacokinetic and pharmacodynamic effects than insulin aspart. Diabetes Obes Metab. 2015 Jul;17(7):682–8.
  36. Rave K, Bott S, Heinemann L, Sha S, Becker RHA, Willavize SA, et al. Time-Action Profile of Inhaled Insulin in Comparison With Subcutaneously Injected Insulin Lispro and Regular Human Insulin. Diabetes Care. 2005 May 1;28(5):1077–82.
  37. Biester T, Kordonouri O, Danne T. Pharmacological Properties of Faster-Acting Insulin Aspart. Curr Diab Rep. 2017 Sep 23;17(11):101.
  38. Plank J, Bodenlenz M, Sinner F, Magnes C, Görzer E, Regittnig W, et al. A Double-Blind, Randomized, Dose-Response Study Investigating the Pharmacodynamic and Pharmacokinetic Properties of the Long-Acting Insulin Analog Detemir. Diabetes Care. 2005 May 1;28(5):1107–12.
  39. Lepore M, Pampanelli S, Fanelli C, Porcellati F, Bartocci L, Di Vincenzo A, et al. Pharmacokinetics and pharmacodynamics of subcutaneous injection of long-acting human insulin analog glargine, NPH insulin, and ultralente human insulin and continuous subcutaneous infusion of insulin lispro. Diabetes. 2000 Dec 1;49(12):2142–8.
  40. Rifkin H, Sherwin RS, Porte D, Ellenberg M, editors. Ellenberg & Rifkin’s diabetes mellitus. 5th ed. /. Stamford, Conn. : Appleton & Lange,; c1997.
  41. Lalli C, Ciofetta M, Del Sindaco P, Torlone E, Pampanelli S, Compagnucci P, et al. Long-term intensive treatment of type 1 diabetes with the short-acting insulin analog lispro in variable combination with NPH insulin at mealtime. Diabetes Care. 1999 Mar 1;22(3):468–77.
  42. Brunelle RL, Llewelyn J, Anderson JH Jr, Gale EAM, Koivisto VA. Meta-Analysis of the Effect of Insulin Lispro on Severe Hypoglycemia in Patients With Type 1 Diabetes. Diabetes Care. 1998 Oct 1;21(10):1726–31.
  43. Pampanelli S, Torlone E, Ialli C, Sindaco PD, Ciofetta M, Lepore M, et al. Improved Postprandial Metabolic Control After Subcutaneous Injection of a Short-Acting Insulin Analog in IDDM of Short Duration With Residual Pancreatic β-Cell Function. Diabetes Care. 1995 Nov 1;18(11):1452–9.
  44. Cobry E, McFann K, Messer L, Gage V, VanderWel B, Horton L, et al. Timing of Meal Insulin Boluses to Achieve Optimal Postprandial Glycemic Control in Patients with Type 1 Diabetes. Diabetes Technol Ther. 2010 Mar;12(3):173–7.
  45. Luijf YM, Bon AC van, Hoekstra JB, DeVries JH. Premeal Injection of Rapid-Acting Insulin Reduces Postprandial Glycemic Excursions in Type 1 Diabetes. Diabetes Care. 2010 Oct;33(10):2152.
  46. Klonoff DC, Evans ML, Lane W, Kempe HP, Renard E, DeVries JH, et al. A randomized, multicentre trial evaluating the efficacy and safety of fast-acting insulin aspart in continuous subcutaneous insulin infusion in adults with type 1 diabetes (onset 5). Diabetes Obes Metab. 2019;21(4):961–7.
  47. Warren M, Bode B, Cho JI, Liu R, Tobian J, Hardy T, et al. Improved postprandial glucose control with ultra rapid lispro versus lispro with continuous subcutaneous insulin infusion in type 1 diabetes: PRONTO-Pump-2. Diabetes Obes Metab. 2021;23(7):1552–61.
  48. Important Safety Information | Insulin Pump Therapy | Omnipod [Internet]. [cited 2023 Feb 12]. Available from: https://www.omnipod.com/safety
  49. Insulin Compatibility [Internet]. Support. [cited 2023 Feb 5]. Available from: https://support.tandemdiabetes.com/hc/en-us/articles/1500011397082-Insulin-Compatibility
  50. Medtronic Diabetes Useful Information [Internet]. Medtronic Diabetes. 2012 [cited 2023 Feb 12]. Available from: https://www.medtronicdiabetes.com/search
  51. Tupola S, Komulainen J, Jääskeläinen J, Sipilä I. Post-prandial insulin lispro vs. human regular insulin in prepubertal children with Type 1 diabetes mellitus. Diabet Med J Br Diabet Assoc. 2001 Aug;18(8):654–8.
  52. Kumar D. Lispro Analog for Treatment of Generalized Allergy to Human Insulin. Diabetes Care. 1997 Sep 1;20(9):1357–9.
  53. Heise T, Hövelmann U, Zijlstra E, Stender-Petersen K, Jacobsen JB, Haahr H. A Comparison of Pharmacokinetic and Pharmacodynamic Properties Between Faster-Acting Insulin Aspart and Insulin Aspart in Elderly Subjects with Type 1 Diabetes Mellitus. Drugs Aging. 2017;34(1):29–38.
  54. Kildegaard J, Buckley ST, Nielsen RH, Povlsen GK, Seested T, Ribel U, et al. Elucidating the Mechanism of Absorption of Fast-Acting Insulin Aspart: The Role of Niacinamide. Pharm Res. 2019;36(3):49.
  55. Rave K, Heise T, Heinemann L, Boss AH. Inhaled Technosphere® Insulin in Comparison to Subcutaneous Regular Human Insulin: Time Action Profile and Variability in Subjects with Type 2 Diabetes. J Diabetes Sci Technol. 2008 Mar;2(2):205.
  56. Coverage of Postprandial Blood Glucose Excursions With Inhaled Technosphere Insulin in Comparison to Subcutaneously Injected Regular Human Insulin in Subjects With Type 2 Diabetes | Diabetes Care | American Diabetes Association [Internet]. [cited 2022 Dec 28]. Available from: https://diabetesjournals.org/care/article/30/9/2307/29333/Coverage-of-Postprandial-Blood-Glucose-Excursions
  57. Rosenstock J, Bergenstal R, DeFronzo RA, Hirsch IB, Klonoff D, Boss AH, et al. Efficacy and Safety of Technosphere Inhaled Insulin Compared With Technosphere Powder Placebo in Insulin-Naive Type 2 Diabetes Suboptimally Controlled With Oral Agents. Diabetes Care. 2008 Nov 1;31(11):2177–82.
  58. Rosenstock J, Lorber DL, Gnudi L, Howard CP, Bilheimer DW, Chang PC, et al. Prandial inhaled insulin plus basal insulin glargine versus twice daily biaspart insulin for type 2 diabetes: a multicentre randomised trial. The Lancet. 2010 Jun 26;375(9733):2244–53.
  59. AFREZZA® (insulin human) Inhalation Powder.
  60. The pharmacokinetics and pharmacodynamics of rapid‐acting insulin analogues and their clinical consequences - Home - 2012 - Diabetes, Obesity and Metabolism - Wiley Online Library [Internet]. [cited 2022 Dec 28]. Available from: https://dom-pubs.onlinelibrary.wiley.com/doi/10.1111/j.1463-1326.2012.01580.x
  61. T H, K SP, U H, Jb J, L N, E Z, et al. Pharmacokinetic and Pharmacodynamic Properties of Faster-Acting Insulin Aspart versus Insulin Aspart Across a Clinically Relevant Dose Range in Subjects with Type 1 Diabetes Mellitus. Clin Pharmacokinet [Internet]. 2017 Jun [cited 2022 Dec 28];56(6). Available from: https://pubmed.ncbi.nlm.nih.gov/27878566/
  62. Heise T, Linnebjerg H, Coutant D, LaBell E, Zijlstra E, Kapitza C, et al. Ultra rapid lispro lowers postprandial glucose and more closely matches normal physiological glucose response compared to other rapid insulin analogues: A phase 1 randomized, crossover study. Diabetes Obes Metab. 2020;22(10):1789–98.
  63. Bolli GB, Owens DR. Insulin glargine. Lancet Lond Engl. 2000 Aug 5;356(9228):443–5.
  64. Heinemann L, Linkeschova R, Rave K, Hompesch B, Sedlak M, Heise T. Time-action profile of the long-acting insulin analog insulin glargine (HOE901) in comparison with those of NPH insulin and placebo. Diabetes Care. 2000 May 1;23(5):644–9.
  65. Mudaliar S, Mohideen P, Deutsch R, Ciaraldi TP, Armstrong D, Kim B, et al. Intravenous Glargine and Regular Insulin Have Similar Effects on Endogenous Glucose Output and Peripheral Activation/Deactivation Kinetic Profiles. Diabetes Care. 2002 Sep 1;25(9):1597–602.
  66. Yki-Järvinen H, Dressler A, Ziemen M, HOE 901/300s Study Group. Less nocturnal hypoglycemia and better post-dinner glucose control with bedtime insulin glargine compared with bedtime NPH insulin during insulin combination therapy in type 2 diabetes. HOE 901/3002 Study Group. Diabetes Care. 2000 Aug 1;23(8):1130–6.
  67. Ratner RE, Hirsch IB, Neifing JL, Garg SK, Mecca TE, Wilson CA. Less hypoglycemia with insulin glargine in intensive insulin therapy for type 1 diabetes. U.S. Study Group of Insulin Glargine in Type 1 Diabetes. Diabetes Care. 2000 May 1;23(5):639–43.
  68. Heise T, Hermanski L, Nosek L, Feldman A, Rasmussen S, Haahr H. Insulin degludec: four times lower pharmacodynamic variability than insulin glargine under steady-state conditions in type 1 diabetes. Diabetes Obes Metab. 2012;14(9):859–64.
  69. Ratner RE, Gough SCL, Mathieu C, Prato SD, Bode B, Mersebach H, et al. Hypoglycaemia risk with insulin degludec compared with insulin glargine in type 2 and type 1 diabetes: a pre-planned meta-analysis of phase 3 trials. Diabetes Obes Metab. 2013 Feb;15(2):175.
  70. Marso SP, McGuire DK, Zinman B, Poulter NR, Emerson SS, Pieber TR, et al. Efficacy and Safety of Degludec versus Glargine in Type 2 Diabetes. N Engl J Med. 2017 Aug 24;377(8):723–32.
  71. Effect of Insulin Degludec vs Insulin Glargine U100 on Hypoglycemia in Patients With Type 1 Diabetes: The SWITCH 1 Randomized Clinical Trial | Clinical Pharmacy and Pharmacology | JAMA | JAMA Network [Internet]. [cited 2022 Dec 28]. Available from: https://jamanetwork.com/journals/jama/fullarticle/2635629
  72. Wysham C, Bhargava A, Chaykin L, de la Rosa R, Handelsman Y, Troelsen LN, et al. Effect of Insulin Degludec vs Insulin Glargine U100 on Hypoglycemia in Patients With Type 2 Diabetes: The SWITCH 2 Randomized Clinical Trial. JAMA. 2017 Jul 4;318(1):45–56.
  73. Grunberger G. Insulin Analogs—Are They Worth It? Yes! Diabetes Care. 2014 May 10;37(6):1767–70.
  74. Riddle MC, Rosenstock J, Gerich J, on behalf of the Insulin Glargine 4002 Study Investigators. The Treat-to-Target Trial: Randomized addition of glargine or human NPH insulin to oral therapy of type 2 diabetic patients. Diabetes Care. 2003 Nov 1;26(11):3080–6.
  75. Vague P, Selam JL, Skeie S, De Leeuw I, Elte JWF, Haahr H, et al. Insulin Detemir Is Associated With More Predictable Glycemic Control and Reduced Risk of Hypoglycemia Than NPH Insulin in Patients With Type 1 Diabetes on a Basal-Bolus Regimen With Premeal Insulin Aspart. Diabetes Care. 2003 Mar 1;26(3):590–6.
  76. Riddle MC, Bolli GB, Ziemen M, Muehlen-Bartmer I, Bizet F, Home PD, et al. New Insulin Glargine 300 Units/mL Versus Glargine 100 Units/mL in People With Type 2 Diabetes Using Basal and Mealtime Insulin: Glucose Control and Hypoglycemia in a 6-Month Randomized Controlled Trial (EDITION 1). Diabetes Care. 2014 Sep 10;37(10):2755–62.
  77. Yki-Järvinen H, Bergenstal R, Ziemen M, Wardecki M, Muehlen-Bartmer I, Boelle E, et al. New Insulin Glargine 300 Units/mL Versus Glargine 100 Units/mL in People With Type 2 Diabetes Using Oral Agents and Basal Insulin: Glucose Control and Hypoglycemia in a 6-Month Randomized Controlled Trial (EDITION 2). Diabetes Care. 2014 Nov 8;37(12):3235–43.
  78. Bolli GB, Riddle MC, Bergenstal RM, Ziemen M, Sestakauskas K, Goyeau H, et al. New insulin glargine 300 U/ml compared with glargine 100 U/ml in insulin-naïve people with type 2 diabetes on oral glucose-lowering drugs: a randomized controlled trial (EDITION 3). Diabetes Obes Metab. 2015;17(4):386–94.
  79. Holman RR, Farmer AJ, Davies MJ, Levy JC, Darbyshire JL, Keenan JF, et al. Three-Year Efficacy of Complex Insulin Regimens in Type 2 Diabetes. N Engl J Med. 2009 Oct 29;361(18):1736–47.
  80. Newsome C. Basaglar. Clin Diabetes Publ Am Diabetes Assoc. 2017 Jul;35(3):181.
  81. Commissioner O of the. FDA approves Admelog, the first short-acting “follow-on” insulin product to treat diabetes [Internet]. FDA. FDA; 2020 [cited 2022 Dec 27]. Available from: https://www.fda.gov/news-events/press-announcements/fda-approves-admelog-first-short-acting-follow-insulin-product-treat-diabetes
  82. Sun B, Sengupta N, Rao A, Donnelly C, Waichale V, Roy AS, et al. Similar immunogenicity profiles between the proposed biosimilar MYL-1501D and reference insulin glargine in patients with diabetes mellitus: the phase 3 INSTRIDE 1 and INSTRIDE 2 studies. BMC Endocr Disord. 2021 Jun 26;21(1):129.
  83. Blevins TC, Barve A, Sun B, Raiter Y, Aubonnet P, Muniz R, et al. Efficacy and safety of MYL-1501D versus insulin glargine in patients with type 2 diabetes after 24 weeks: Results of the phase III INSTRIDE 2 study. Diabetes Obes Metab. 2019;21(1):129–35.
  84. Blevins TC, Barve A, Raiter Y, Aubonnet P, Athalye S, Sun B, et al. Efficacy and safety of MYL-1501D versus insulin glargine in people with type 1 diabetes mellitus: Results of the INSTRIDE 3 phase 3 switch study. Diabetes Obes Metab. 2020;22(3):365–72.
  85. Cryer PE. Mechanisms of Hypoglycemia-Associated Autonomic Failure in Diabetes. N Engl J Med. 2013 Jul 25;369(4):362–72.
  86. The Effect of Intensive Treatment of Diabetes on the Development and Progression of Long-Term Complications in Insulin-Dependent Diabetes Mellitus. N Engl J Med. 1993 Sep 30;329(14):977–86.
  87. Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). The Lancet. 1998 Sep 12;352(9131):837–53.
  88. Effects of Intensive Glucose Lowering in Type 2 Diabetes | NEJM [Internet]. [cited 2022 Dec 25]. Available from: https://www.nejm.org/doi/10.1056/NEJMoa0802743?url_ver=Z39.88-2003&rfr_id=ori:rid:crossref.org&rfr_dat=cr_pub%20%200www.ncbi.nlm.nih.gov
  89. Intensive Blood Glucose Control and Vascular Outcomes in Patients with Type 2 Diabetes. N Engl J Med. 2008 Jun 12;358(24):2560–72.
  90. Seaquist ER, Anderson J, Childs B, Cryer P, Dagogo-Jack S, Fish L, et al. Hypoglycemia and Diabetes: A Report of a Workgroup of the American Diabetes Association and The Endocrine Society. Diabetes Care. 2013 Apr 13;36(5):1384–95.
  91. McCoy RG, Lipska KJ, Yao X, Ross JS, Montori VM, Shah ND. Intensive Treatment and Severe Hypoglycemia Among Adults With Type 2 Diabetes. JAMA Intern Med. 2016 Jul 1;176(7):969–78.
  92. Desouza C, Salazar H, Cheong B, Murgo J, Fonseca V. Association of Hypoglycemia and Cardiac Ischemia: A study based on continuous monitoring. Diabetes Care. 2003 May 1;26(5):1485–9.
  93. Frier BM, Schernthaner G, Heller SR. Hypoglycemia and Cardiovascular Risks. Diabetes Care. 2011 May;34(Suppl 2):S132–7.
  94. Donner T, Muñoz M. Update on Insulin Therapy for Type 2 Diabetes. J Clin Endocrinol Metab. 2012 May 1;97(5):1405–13.
  95. Miller ME, Bonds DE, Gerstein HC, Seaquist ER, Bergenstal RM, Calles-Escandon J, et al. The effects of baseline characteristics, glycaemia treatment approach, and glycated haemoglobin concentration on the risk of severe hypoglycaemia: post hoc epidemiological analysis of the ACCORD study. BMJ. 2010 Jan 8;340:b5444.
  96. Bremer JP, Jauch-Chara K, Hallschmid M, Schmid S, Schultes B. Hypoglycemia Unawareness in Older Compared With Middle-Aged Patients With Type 2 Diabetes. Diabetes Care. 2009 Aug;32(8):1513–7.
  97. de Galan BE, Zoungas S, Chalmers J, Anderson C, Dufouil C, Pillai A, et al. Cognitive function and risks of cardiovascular disease and hypoglycaemia in patients with type 2 diabetes: the Action in Diabetes and Vascular Disease: Preterax and Diamicron Modified Release Controlled Evaluation (ADVANCE) trial. Diabetologia. 2009 Nov 1;52(11):2328–36.
  98. ElSayed NA, Aleppo G, Aroda VR, Bannuru RR, Brown FM, Bruemmer D, et al. 6. Glycemic Targets: Standards of Care in Diabetes—2023. Diabetes Care. 2022 Dec 12;46(Supplement_1):S97–110.
  99. Laffel LM, Kanapka LG, Beck RW, Bergamo K, Clements MA, Criego A, et al. Effect of Continuous Glucose Monitoring on Glycemic Control in Adolescents and Young Adults With Type 1 Diabetes. JAMA. 2020 Jun 16;323(23):2388–96.
  100. Pratley RE, Kanapka LG, Rickels MR, Ahmann A, Aleppo G, Beck R, et al. Effect of Continuous Glucose Monitoring on Hypoglycemia in Older Adults With Type 1 Diabetes. JAMA. 2020 Jun 16;323(23):2397–406.
  101. Meneghini L, Atkin SL, Gough SCL, Raz I, Blonde L, Shestakova M, et al. The Efficacy and Safety of Insulin Degludec Given in Variable Once-Daily Dosing Intervals Compared With Insulin Glargine and Insulin Degludec Dosed at the Same Time Daily: A 26-week, randomized, open-label, parallel-group, treat-to-target trial in individuals with type 2 diabetes. Diabetes Care. 2013 Mar 14;36(4):858–64.
  102. Rosenstock J, Davies M, Home PD, Larsen J, Koenen C, Schernthaner G. A randomised, 52-week, treat-to-target trial comparing insulin detemir with insulin glargine when administered as add-on to glucose-lowering drugs in insulin-naive people with type 2 diabetes. Diabetologia. 2008 Mar;51(3):408–16.
  103. Davies MJ, Derezinski T, Pedersen CB, Clauson P. Reduced Weight Gain with Insulin Detemir Compared to NPH Insulin Is Not Explained by a Reduction in Hypoglycemia. Diabetes Technol Ther. 2008 Aug;10(4):273–7.
  104. Buse JB, Wolffenbuttel BHR, Herman WH, Hippler S, Martin SA, Jiang HH, et al. The DURAbility of Basal versus Lispro mix 75/25 insulin Efficacy (DURABLE) Trial. Diabetes Care. 2011 Feb;34(2):249–55.
  105. Radermecker RP, Scheen AJ. Allergy reactions to insulin: effects of continuous subcutaneous insulin infusion and insulin analogues. Diabetes Metab Res Rev. 2007;23(5):348–55.
  106. Babiker A, Datta V. Lipoatrophy with insulin analogues in type I diabetes. Arch Dis Child. 2011 Jan 1;96(1):101–2.
  107. Colhoun HM. Use of insulin glargine and cancer incidence in Scotland: a study from the Scottish Diabetes Research Network Epidemiology Group. Diabetologia. 2009 Sep;52(9):1755–65.
  108. Jonasson JM, Ljung R, Talbäck M, Haglund B, Gudbjörnsdòttir S, Steineck G. Insulin glargine use and short-term incidence of malignancies—a population-based follow-up study in Sweden. Diabetologia. 2009 Sep 1;52(9):1745–54.
  109. Hemkens LG, Grouven U, Bender R, Günster C, Gutschmidt S, Selke GW, et al. Risk of malignancies in patients with diabetes treated with human insulin or insulin analogues: a cohort study. Diabetologia. 2009 Sep;52(9):1732–44.
  110. Home PD, Lagarenne P. Combined randomised controlled trial experience of malignancies in studies using insulin glargine. Diabetologia. 2009;52(12):2499–506.
  111. Basal Insulin and Cardiovascular and Other Outcomes in Dysglycemia | NEJM [Internet]. [cited 2022 Dec 25]. Available from: https://www.nejm.org/doi/full/10.1056/nejmoa1203858
  112. Schroeder EB, Neugebauer R, Reynolds K, Schmittdiel JA, Loes L, Dyer W, et al. Association of Cardiovascular Outcomes and Mortality With Sustained Long-Acting Insulin Only vs Long-Acting Plus Short-Acting Insulin Treatment. JAMA Netw Open. 2021 Sep 24;4(9):e2126605.
  113. Hirsch IB. Type 1 Diabetes Mellitus and the Use of Flexible Insulin Regimens. Am Fam Physician. 1999 Nov 15;60(8):2343–52.
  114. Low Wang CC, Shah AC. Medical Management of Type 1 Diabetes [Internet]. American Diabetes Association; 2017 [cited 2022 Dec 19]. Available from: https://doi.org/10.2337/9781580406307
  115. Chiang JL, Kirkman MS, Laffel LMB, Peters AL, on behalf of the Type 1 Diabetes Sourcebook Authors. Type 1 Diabetes Through the Life Span: A Position Statement of the American Diabetes Association. Diabetes Care. 2014 Jun 12;37(7):2034–54.
  116. Intensive Diabetes Treatment and Cardiovascular Disease in Patients with Type 1 Diabetes. N Engl J Med. 2005 Dec 22;353(25):2643–53.
  117. Nathan DM, Buse JB, Davidson MB, Ferrannini E, Holman RR, Sherwin R, et al. Medical Management of Hyperglycemia in Type 2 Diabetes: A Consensus Algorithm for the Initiation and Adjustment of Therapy: A consensus statement of the American Diabetes Association and the European Association for the Study of Diabetes. Diabetes Care. 2009 Jan 1;32(1):193–203.
  118. Hermann LS. Optimising Therapy for Insulin-Treated Type 2 Diabetes Mellitus. Drugs Aging. 2000 Oct 1;17(4):283–94.
  119. DeWitt DE, Dugdale DC. Using New Insulin Strategies in the Outpatient Treatment of DiabetesClinical Applications. JAMA. 2003 May 7;289(17):2265–9.
  120. Fanelli CG, Pampanelli S, Porcellati F, Rossetti P, Brunetti P, Bolli GB. Administration of Neutral Protamine Hagedorn Insulin at Bedtime versus with Dinner in Type 1 Diabetes Mellitus To Avoid Nocturnal Hypoglycemia and Improve Control. Ann Intern Med. 2002 Apr 2;136(7):504–14.
  121. Francis AJ, Home PD, Hanning I, Alberti KG, Tunbridge WM. Intermediate acting insulin given at bedtime: effect on blood glucose concentrations before and after breakfast. Br Med J Clin Res Ed. 1983 Apr 9;286(6372):1173–6.
  122. Raskin P, Bode BW, Marks JB, Hirsch IB, Weinstein RL, McGill JB, et al. Continuous Subcutaneous Insulin Infusion and Multiple Daily Injection Therapy Are Equally Effective in Type 2 Diabetes : A randomized, parallel-group, 24-week study. Diabetes Care. 2003 Sep 1;26(9):2598–603.
  123. Mudaliar S, Edelman SV. INSULIN THERAPY IN TYPE 2 DIABETES. Endocrinol Metab Clin North Am. 2001 Dec 1;30(4):935–82.
  124. Bode BW, Sabbah HT, Gross TM, Fredrickson LP, Davidson PC. Diabetes management in the new millennium using insulin pump therapy. Diabetes Metab Res Rev. 2002;18(S1):S14–20.
  125. Bode BW, Tamborlane WV, Davidson PC. Insulin pump therapy in the 21st century: Strategies for successful use in adults, adolescents, and children with diabetes. Postgrad Med. 2002 May;111(5):69–77.
  126. Pickup J, Keen H. Continuous Subcutaneous Insulin Infusion at 25 Years: Evidence base for the expanding use of insulin pump therapy in type 1 diabetes. Diabetes Care. 2002 Mar 1;25(3):593–8.
  127. Brown SA, Kovatchev BP, Raghinaru D, Lum JW, Buckingham BA, Kudva YC, et al. Six-Month Randomized, Multicenter Trial of Closed-Loop Control in Type 1 Diabetes. N Engl J Med. 2019 Oct 31;381(18):1707–17.
  128. Weaver KW, Hirsch IB. The Hybrid Closed-Loop System: Evolution and Practical Applications. Diabetes Technol Ther. 2018 Jun;20(S2):S2-16.
  129. Breton MD, Kovatchev BP. One Year Real-World Use of the Control-IQ Advanced Hybrid Closed-Loop Technology. Diabetes Technol Ther. 2021 Sep;23(9):601–8.
  130. Berget C, Sherr JL, DeSalvo DJ, Kingman RS, Stone SL, Brown SA, et al. Clinical Implementation of the Omnipod 5 Automated Insulin Delivery System: Key Considerations for Training and Onboarding People With Diabetes. Clin Diabetes. 2022 Apr 15;40(2):168–84.
  131. Zhou K, Isaacs D. Closed-Loop Artificial Pancreas Therapy for Type 1 Diabetes. Curr Cardiol Rep. 2022 Sep 1;24(9):1159–67.
  132. Blonde L, Merilainen M, Karwe V, Raskin P, Group TS. Patient-directed titration for achieving glycaemic goals using a once-daily basal insulin analogue: an assessment of two different fasting plasma glucose targets - the TITRATETM study. Diabetes Obes Metab. 2009;11(6):623–31.
  133. Holleman F, Van Den Brand JJ, Hoven RA, Van Der Linden JM, Van Der Tweel I, Hoekstra JBL, et al. Comparison of LysB28, ProB29-Human Insulin Analog and Regular Human Insulin in the Correction of Incidental Hyperglycemia. Diabetes Care. 1996 Dec 1;19(12):1426–9.
  134. Kong N, Kitchen MM, Ryder RE. The use of lispro for high sugar content snacks between meals in intensive insulin regimens. Diabet Med J Br Diabet Assoc. 2000 Apr;17(4):331–2.
  135. Training in flexible, intensive insulin management to enable dietary freedom in people with type 1 diabetes: dose adjustment for normal eating (DAFNE) randomised controlled trial. BMJ. 2002 Oct 5;325(7367):746.
  136. Hirsch IB, Farkas-Hirsch R, Skyler JS. Intensive Insulin Therapy for Treatment of Type I Diabetes. Diabetes Care. 1990 Dec 1;13(12):1265–83.
  137. Riddell MC, Gallen IW, Smart CE, Taplin CE, Adolfsson P, Lumb AN, et al. Exercise management in type 1 diabetes: a consensus statement. Lancet Diabetes Endocrinol. 2017 May 1;5(5):377–90.
  138. Moser O, Riddell MC, Eckstein ML, Adolfsson P, Rabasa-Lhoret R, van den Boom L, et al. Glucose management for exercise using continuous glucose monitoring (CGM) and intermittently scanned CGM (isCGM) systems in type 1 diabetes: position statement of the European Association for the Study of Diabetes (EASD) and of the International Society for Pediatric and Adolescent Diabetes (ISPAD) endorsed by JDRF and supported by the American Diabetes Association (ADA). Diabetologia. 2020 Dec 1;63(12):2501–20.
  139. Evidence of a Strong Association Between Frequency of Self-Monitoring of Blood Glucose and Hemoglobin A1c Levels in T1D Exchange Clinic Registry Participants | Diabetes Care | American Diabetes Association [Internet]. [cited 2022 Dec 18]. Available from: https://diabetesjournals.org/care/article/36/7/2009/33033/Evidence-of-a-Strong-Association-Between-Frequency
  140. Can capillary whole blood glucose and venous plasma glucose measurements be used interchangeably in diagnosis of diabetes mellitus? [Internet]. [cited 2022 Dec 18]. Available from: https://www.tandfonline.com/doi/epdf/10.1080/003655102753611799?needAccess=true&role=button
  141. Danne T, Nimri R, Battelino T, Bergenstal RM, Close KL, DeVries JH, et al. International Consensus on Use of Continuous Glucose Monitoring. Diabetes Care. 2017 Nov 10;40(12):1631–40.
  142. Haak T, Hanaire H, Ajjan R, Hermanns N, Riveline JP, Rayman G. Flash Glucose-Sensing Technology as a Replacement for Blood Glucose Monitoring for the Management of Insulin-Treated Type 2 Diabetes: a Multicenter, Open-Label Randomized Controlled Trial. Diabetes Ther. 2017 Feb;8(1):55–73.
  143. Bolinder J, Antuna R, Geelhoed-Duijvestijn P, Kröger J, Weitgasser R. Novel glucose-sensing technology and hypoglycaemia in type 1 diabetes: a multicentre, non-masked, randomised controlled trial. The Lancet. 2016 Nov 5;388(10057):2254–63.
  144. Continuous glucose monitoring in people with diabetes: the randomized controlled Glucose Level Awareness in Diabetes Study (GLADIS) - New - 2015 - Diabetic Medicine - Wiley Online Library [Internet]. [cited 2022 Dec 18]. Available from: https://onlinelibrary.wiley.com/doi/10.1111/dme.12713
  145. Glycaemic control in type 1 diabetes during real time continuous glucose monitoring compared with self monitoring of blood glucose: meta-analysis of randomised controlled trials using individual patient data | The BMJ [Internet]. [cited 2022 Dec 18]. Available from: https://www.bmj.com/content/343/bmj.d3805.long
  146. Managing Sick Days | Diabetes | CDC [Internet]. 2022 [cited 2022 Dec 18]. Available from: https://www.cdc.gov/diabetes/managing/flu-sick-days.html

 

 

Lipid Screening in Youth

 ABSTRACT

 

As improvements in cardiovascular disease (CVD) risk reduction in adults’ plateau and risk factors accumulate in youth, focus is shifting to children as the future of CVD prevention. Abnormal lipid levels are relatively common in the pediatric population and treatments are available and effective thereby supporting the need to screen children for abnormal lipids. Recent data suggests that lipid screening is occurring in youth but is neither detecting the expected proportion of affected individuals nor translating into higher rates of therapy. Future work should expand on current screening efforts and overcome identified barriers to lipid screening toward the goal of avoiding CVD events and maintaining the ideal CVD health of childhood throughout the life course.

INTRODUCTION

 

Salutary trends in adult CVD mortality are documented and appear to stem largely from improvements in atherosclerotic risk factor treatment (1). But key danger signals are also present. First the rate of improvement is waning. Second and perhaps not coincidentally, recent advances focus on reducing thresholds for pharmacological risk factor management and thereby enlarge the proportion of the population eligible for therapy (2, 3). Finally, an alarming trend towards high and increasing obesity, obesity-related dyslipidemia, and type 2 diabetes portend an impending tidal wave of CVD (1).

 

Recent data has demonstrated the first continuous decline in average life expectancy during peacetime in modern American history wherein some component is due to increasing ASCVD in older age groups (4). Population data from 1999-2016 demonstrates lipid abnormalities in one-fifth of children and one-fourth of teenagers (5). At a point where progress is plateauing and efforts are being made to medicate wider swaths of the adult population, children offer an opportunity in the life course to further intensify CVD risk reduction. Childhood is a key time point for progress because children are already accumulating atherosclerotic phenotypic changes, have a high prevalence of CVD risk factors, are susceptible to deleterious lifestyle influences but are also malleable to lifestyle habit alterations. Typically, children have not yet suffered from actual CVD events nor are they likely to in youth. As CVD primary prevention is preventing the first CVD event by the treatment of risk factors, and secondary prevention is evading recurrent CVD events in patients with a history of CVD, primordial prevention aims to prevent or delay development of CVD risk factors.

 

Professional groups including the American Academy of Pediatrics, American Diabetes Association and governmental entities including the National Heart, Lung, and Blood Institute (NHLBI), Department of Health and Human Services have promulgated scientific statements and practice preferences identifying primordial and primary CVD prevention generally and dyslipidemia management specifically as priority area (6-8). The 2011 NHLBI guidelines recommend universal lipid screening for the general population at age 9-11 years. The most recent American Heart Association (AHA) guidelines on CVD risk reduction in high-risk pediatric patients including homozygous FH, type 1 and 2 diabetes, end-stage renal disease, KD with persistent aneurysms, solid organ transplant vasculopathy, and childhood cancer survivors recommends non-fasting non-HDL screening yearly (9).

 

It is clear that population-wide interventions can be successful, as illustrated by cigarette use reduction (1). Tobacco smoking reduction has been achieved through mobilizing public sentiment; placing restrictions on the procurement, advertisement, and use of tobacco products; and use of economic disincentives. Similar efforts to reduce the causes of hyperlipidemia, hypertension, or obesity in adults meet entrenched resistance from the lack of data supporting second-hand harm from these lifestyle behaviors leading to trepidation about restricting an individual’s freedom of personal choice. In contrast, addressing CVD risk factors in children may be more acceptable because their lifestyle choices are appropriately constrained by caregivers. To illustrate, the fact a child would consistently choose ice cream over cauliflower every day is immaterial to whether daily ice cream consumption in children should be discouraged. Therefore, focusing on children offers an opportunity to leverage an identified CVD risk factor abnormality into a multifaceted cardiometabolic remedy. Moreover, children are a powerful motivating factor for lifestyle change in their parents offering the promise for a multiplicative effect on a pediatric intervention. But first we must find affected children.

WHY IS PEDIATRIC LIPID SCREENING APPROPRIATE?

 

The passionate pediatric provider might be motivated to identify all CVD risk factors in every child with the hopes of improving the health of the population one individual at a time. But from a policy and implementation perspective screening tests entail certain trade-offs that must be addressed. These pitfalls include the occurrence of false testing results that may be rare in any individual case but virtually guaranteed when mandatorily applied to many cases; the downstream effects of false test results in terms of additional confirmatory testing and patient emotional distress; test-related harms when instantiated widely; ethical conflicts between identifying sick individuals versus testing related physical and emotional harms to unaffected individuals; and lastly cost-effectiveness concerns. Each of these general concerns is amplified when the patient in question is a developing, vulnerable child for whom identifying risk factors has lasting implications but screening related harms can also have lasting implication rippling through the family. To be more specific, whereas adult providers find a patient blood draw to be trivial, violating bodily integrity is not as facile in children or for their parents, and therefore for providers to order. Nonetheless, many diseases are screened for including with blood testing in the extremely vulnerable newborn period (10). This state screening of newborns searches for disorders with prevalence’s on the order of 0.02% for sickle cell disease to 0.004% for phenylketonuria. Each of the screened disorders has therapies of varying efficacy by disease. Decisions to screen for these diseases are in some part determined by adherence to the World Health Organization Criteria for screening after Wilson and Jungner’s classic formulation (Table 1) (11). These classic criteria offer excellent structure for a discussion of pediatric lipid screening.

 

TABLE 1. Wilson & Jungner Criteria (11)

1. The condition sought should be an important health problem.

2. There should be an accepted treatment for patients with recognized disease.

3. Facilities for diagnosis and treatment should be available.

4. There should be a recognizable latent or early symptomatic stage.

5. There should be a suitable test or examination.

6. The test should be acceptable to the population.

7. The natural history of the condition, including development from latent to declared disease, should be adequately understood.

8. There should be an agreed policy on whom to treat as patients.

9. The cost of case-finding (including diagnosis and treatment of patients diagnosed) should be economically balanced in relation to possible expenditure on medical care as a whole.

10. Case-finding should be a continuing process and not a “once and for all” project.

 

NATURAL HISTORY, LATENCY, IMPORTANCE

 

The causal relation between low density lipoprotein cholesterol (LDL) and CVD events is well established (12-17). Interventions of triglycerides (TG) have not been quite as successful but observational studies using genes in instrumental analysis have determined a prospective unconfounded relation between TG and CVD events (18,19). High density lipoprotein cholesterol (HDL) is associated with incident CVD in observational cohorts but multiple HDL specific interventions have not led to CVD event reduction leading to doubts about the so-called HDL hypothesis (16, 20-22).

 

In general, the relationship between lipid disorders and CVD events is well established. In children it is especially well studied in the Mendelian genetic disorder familial hypercholesterolemia (FH), generally attributed to a dominant negative mutation in the receptor for LDL receptor or apolipoprotein B component of LDL (13). Heterozygous FH occurs in 1 in 500 births while more recent studies suggest it may be as common as 1 in 250. Homozygous FH may be as common as 1 in 160,000 to 1 million (23,24). FH leads to markedly elevated LDL levels. Heterozygous girls suffer coronary events before age 60 in approximately 20% of cases and boys in 50% of cases, while homozygous children have events in the second decade of life (25-27,24). Prior to these events, these children are well documented to have vascular changes predictive of future CVD events (28). Even more common is lifestyle related high TG, low HDL atherogenic dyslipidemia which is present in nearly one in five youth under 17 years old (29,30). Data from young adults in CVD-free general population who have suffered unfortunate mortality from unrelated causes clearly demonstrate arterial atherosclerotic plaques and these plaques are predicted by elevated lipid levels earlier in life (31-33). Lipids predict thicker carotid intimal medial thickness, stiffer aorta, and other preclinical atherosclerotic changes in CVD unaffected individuals (28, 34-37). More recent data combining multiple youth cohorts into a single meta-cohort followed through adulthood clearly demonstrates lipids in childhood directly predict clinically definitive “hard” adult CVD events. Intriguing data also suggests childhood lipoprotein (a) concentrations predict adult “hard” CVD events, including in combination with other CVD risk factors like lipids. Thus, severe and moderate lipid abnormalities occur in youth; these dyslipidemias and hyperlipidemias have important consequences following a predictable pattern from lipid elevation to atherosclerotic progression and eventually CVD events; and are orders of magnitude more common than already universally screened for metabolic conditions.

ACCURATE, SUITABLE, FACILE, REPEATABLE TESTING  

Blood testing is the definitive, rather straightforward mode of lipid abnormality detection with false positive rates of less than one percent. Classic practice is to obtain fasting lipid panels as the ideal, especially for detection of triglyceride elevations (6,27,38). However, obtaining fasting lipids in children can be challenging and so non-fasting lipid panels may be obtained initially with fasting panels obtained to confirm as necessary in an attempt to enhance acceptability (6). In addition, life-course issues are a core concern in lipid assessment of children. While prenatal detection of dyslipidemia is noted, infancy and young childhood is a notoriously difficult period for dyslipidemia assessment due to wildly varied diet habits and food preferences during a child’s introduction and embrace of solid food intake. Toddlers not infrequently habituate to an extremely limited dietary range which they broaden a few years later. Dietary habits and lipid levels tend to stabilize in the early school age until around 10 years of age when pubertal changes with a high degree of variability. Hormonal changes around puberty can be associated with substantial changes in lipid levels (6,25,38). Thus, children could be inappropriately labeled “abnormal” from lipid tests since CVD risk factors fluctuate throughout childhood and adulthood (39). The NHLBI Integrated Guidelines for CVD Risk Reduction in Children and Adolescents recommend taking the average of multiple lipid values to help avoid misclassification and errors from regression to the mean (6). But also similar to adults, single lipid measurements in childhood do predict adult atherosclerotic progression, thereby underscoring the utility of even a single lipid test (31-35).

 

Physical exam findings can induce lipid testing. For example, the presence of tendinous xanthomata on extensor surfaces in young child should trigger lipid investigation for familial hypercholesterolemia or other lipid disorders (6,39). Similarly, many providers appreciate a higher relative risk of lipid abnormalities in overweight individuals. Overweight youth are known to have roughly double the risk of lipid problems while obese youth have roughly three times the risk (29,30,40). However, nearly 10% of normal weight individuals have abnormal lipid levels. So, while it is true that excess weigh individuals are at higher risk of abnormal lipids, the converse is also true, that a substantial proportion of youth with abnormal lipids are normal weight. In fact, since nearly 35-45% of youth with abnormal lipids are normal weight, fixating on excess weight youth misses a substantial proportion of the population’s lipid problem. While the origins of both abnormal lipids and obesity derive from suboptimal diets, activity and inactivity levels, the two are not synonymous. This epidemiological conundrum is a key pillar in favor of the NHLBI guidelines recommending the extension from selective to universal screening of youth depending on age group (Table 2).

 

TABLE 2. NHLBI Recommendations on Lipid Testing by Age Group (6)

Birth to 2 years

No screening

 

2 to 8 years

Selective Screening

Fasting Lipid Profiles (Average of two sets) for:

1st or 2nd degree relative with history of CVD or history of total cholesterol ≥ 240 mg/dL or child has CVD high or moderate risk factors or conditions 

 

9 to 11 years

Universal Screening

Non-fasting Lipid Profile followed by Fasting Lipid Profile for non-HDL≥145 or HDL≤40 or

Fasting Lipid Profile with repeat if LDL≥ 130 mg/dL or non-HDL ≥145 mg/dL or HDL < 40mg/dL or TG ≥100mg/dL for under 10-year-olds; LDL ≥130 mg/dL for at or over 10-year-olds

 

12 to 16 years

Selective Screening

Fasting Lipid Profiles (Average of two sets) for:

1st or 2nd degree relative with history of CVD or history of total cholesterol ≥ 240 mg/dL or child has CVD high or moderate risk factors or conditions

 

17 to 19 years

Universal Screening

Non-Fasting Lipid Profile followed by Fasting Lipid Profile (average two sets) if non–HDL>145 mg/dL or HDL cholesterol< 40 mg/dL or Fasting Lipid Profile and If LDL> 130 mg/dL or non–HDL> 145 mg/dL or HDL< 40 mg/dL or TG> 130 mg/dL. Repeat FLP and average results 

 

20 to 21 years

Universal Screening

Non-Fasting Lipid Profile

Non–HDL> 190 mg/dL or HDL< 40 mg/dL

Measure FLP twice, average results or 

Fasting Lipid Profile

If LDL> 160 mg/dL or non–HDL> 190 mg/dL or HDL< 40 mg/dL or TG> 150 mg/dL

Repeat and average results

CVD: MI, angina, stroke, coronary bypass surgery, coronary stent, coronary angioplasty at or under 55 y in males, 65 y in females 

High risk factors: Hypertension that requires drug therapy (BP> 99th percentile 5 mm Hg), Current cigarette smoker, Body Mass Index at the 97th age-sex specific percentile

High risk conditions: Diabetes mellitus Type 1 or Type 2, Chronic kidney disease, end-stage renal disease, post–renal transplant, post–orthotopic heart transplant, Kawasaki disease with current aneurysms

Moderate risk factors: Hypertension that does not require drug therapy, Body Mass Index between 95th percentile and 97th percentile, HDL< 40 mg/dL

Moderate risk conditions: Kawasaki disease with regressed coronary aneurysms, chronic inflammatory disease (systemic lupus erythematosus, juvenile rheumatoid arthritis), HIV infection, nephrotic syndrome

 

The Guidelines recommend using relatively high thresholds to designate abnormal levels in conjunction with taking the average of multiple lipid values to help avoid misclassification and errors from regression to the mean. The NHLBI guidelines reflect the age-specific distribution of lipid levels while at the same time mirroring the acceptable lipid values category groupings of the Adult Treatment Panel III/National Cholesterol Education Program (Table 3). The key CARDIAC study assessed selective versus universal lipid screening in a general population of more than 20,000 5th graders in West Virginia. Of these more than 70% met NCEP guidelines for selective lipid screening (41). Of those with mildly elevated LDL over 130mg/dL, NCEP guideline based testing did not capture 30% of cases. Of those with LDL at or over 160 mg/dL, NCEP guidelines missed 37% of affected children. Therefore, universal lipid screening identifies children with either a modest or more marked elevations in LDL-C than selective screening. Universal screening becomes an attractive method to detect both genetic and lifestyle related dyslipidemias when considering parental lack of understanding about lipid levels, the ability of lipid lowering medications to prevent CVD events and treat lipid levels in affected parents, or a parent’s refusal to examine their own cholesterol levels hindering screening programs contingent on other exigencies (42-45). The NHLBI guidelines refine the universal screening to apply in age strata around age 10 primarily to detect genetic dyslipidemias and around age 18 when patient-driven lifestyle habits have been established and modifications can still occur just prior to the transition to full adult independence. On balance, lipid disorders appear to be accurately assessed through a simple investigation that can be repeated on multiple occasions.

 

TABLE 3. NHLBI Guideline Lipid Thresholds by Age (mg/dL)

 

Acceptable

Borderline

Abnormal

Total Cholesterol

Children/Adolescents

Young Adults

 

<170

<190

 

170-199

190-224

 

≥200

≥225

LDL Cholesterol

Children/Adolescents

Young Adults

 

<110

<120

 

110-129

120-159

 

≥130

≥160

Non-HDL Cholesterol

Children/Adolescents

Young Adults

 

<120

<150

 

120-144

150-189

 

≥145

≥190

Triglycerides

0-9 years

10-19 years

Young Adults

 

<75

<90

<115

 

75-99

90-129

115-149

 

≥100

≥130

≥150

HDL Cholesterol

Children/Adolescents

Young Adults

 

>45

>45

 

40-45

40-45

 

<40

<40

 

TREATABILITY

 

Lipid disorders also appear to be a treatable phenomenon. Compelling data from Braamskamp et al compared FH offspring who have been treated with statins from an early age followed to age 30 years with their parents until age 30. A dramatic separation in freedom from coronary event curves were seen with cumulative coronary event incidence of 25% in parents while only 1 offspring had an event. The presumed difference between these two genetically comparable groups is the youth age use of 3-hydroxy-3-methyl-glutaryl-CoA reductase (statins). Indeed, the one event in offspring occurred in a youth who self-discontinued therapy. These results suggest long-term LDL-C reduction is beneficial in delaying events (46). That data has recently been extended to show protection through age 40. The effect of statins in treating LDL-C levels has been examined in randomized, placebo-controlled, clinical trials of FH children and found to be safe and efficacious. RCTs in children with FH age 8 to 17 years show those in the statin group had regression of carotid IMT thickness (cIMT) while the placebo group was stable or worsened. In young FH adults, statin use has led to a substantial reduction in coronary mortality (27,28,36,37,47). In the CHARON study children age 6-9, 10-13 and 14-17 treated with rosuvastatin showed LDL level reduction by 43%, 45% and 35% respectively. There were no serious adverse events related to treatment and no deleterious effects on growth or sexual maturation (48). An elegant combined meta-analysis of randomized control trials trial-duration statin therapy was compared to meta-analyzed LDL-lowering genetic mutations on CVD events (49). CVD prevention per unit LDL decrease was several fold more effective by genetic polymorphism than by pharmacologic intervention. The implication was that the degree of LDL lowering was synergistically enhanced by the amount of time spent at a reduced LDL concentration (15,50). Another recent study comparing cholesterol at various ages in adulthood found lowering had better outcomes when occurring earlier in life. (51) On balance observational data abounds on the safety and efficacy of pharmacologic LDL lowering in hyperlipidemia.

 

With respect to dietary modification in LDL-C patients, a key study in pediatric practice was the Dietary Intervention Study in Children which delivered a low total fat, saturated fat, and cholesterol message to 7–10-year-olds with elevated LDL. The trial successfully lowered LDL roughly 10% from baseline (52). The STRIP trial provided similar messaging into the infant age group with similar long-term results and no safety concerns throughout younger childhood (53). While it is true that meta-analytic data from adults suggests that dietary quality alterations are not associated with elevated CVD event risk, broad-based adult cohort studied are inappropriately applied to subpopulations presenting early in life with markedly abnormal lipid values (12). In addition, the NHLBI guidelines pursue primordial prevention by recommending for all children a widely accepted sensible diet approach which moderates simple carbohydrates, processed foods, and saturated fat as well as encourages vegetables and lean proteins.

 

Data on pharmacological or lifestyle modification in youth leading to CVD event reduction in adulthood are not yet available and are unlikely to be forthcoming given the logistical complexity and cost of clinical trials assessing CVD events in large numbers of children over several decades. In the absence of decades long trial data, the data previously quote on life course cholesterol levels are relevant. In addition, anthropological epidemiology demonstrates lower rates of CVD in cultures with habitually low cholesterol on a population basis (15). Additional supportive data comes from cost-effectiveness modeling. Identifying and treating patients with FH yields costs of about $7000/quality-adjusted life year, which generally falls into a willing-to-pay threshold of virtually every high per capita income and many middle per capita income nations (54). Although the additional costs of universal screening are not known, the benefits of earlier CVD prevention in high-risk individuals would be considerable as will cost savings (55). The inferences from lifestyle and pharmacotherapy data stands against a common criticism that youth are not the appropriate population for lipid management. Indeed data, however limited, suggests youth are indeed worthy of respect as persons and health conditions they accrue are also worthy of inspection and intervention.

 

For the highly prevalent, high TG-low HDL so-called atherogenic dyslipidemia, the primary treatment of lifestyle modification has been towards weight management (56-60). These studies have noted consistent relations between weight loss and improved TG and HDL that may persist for up to 5 years. Other data suggests that changes in dietary quality toward a lower carbohydrate intake may be effective in a trend towards TG reduction and HDL improvement. Some publications detail the dominant role of dietary quality recommendations without weight loss documenting a roughly one-third reduction in TG (61). Therefore, specific dietary quality modification can modify abnormal lipids without affecting weight immediately. These dyslipidemia-specific dietary modifications are effective but onerous for families and so should not be applied to the entire population. When motivated to avoid medication, youth and families may become more engaged.

ACCEPTABILITY

 

Focusing on kids ratifies their status as individuals worthy of care independent of their parents. Focusing on kids may also boost identification of dyslipidemic family members in a reverse cascade. Pediatric lipid screening and especially universal screening are controversial despite demonstrated failures of selective screening and examination-based screening (62-64). First, it is highly likely that a very small number will be inappropriately labeled as abnormal lipids due to fluctuating levels during childhood. Second, since obesity increases the risk of abnormal lipid values, objections arise about classifying a multitude of children already psychologically vulnerable from an “abnormal” weight label, with an “abnormal” cholesterol label. Adding to the problems of these already disadvantaged youth makes the child even more demoralized. All providers are concerned about pediatric lipid patients being loosely prescribed statins. The NHLBI panel mandates lifestyle alterations as the primary response, but there is skepticism (64). A survey of US pediatricians in 2013-2014 showed that only 26% were well informed about the 2011 NHLBI guidelines and 68% never or rarely screen healthy 9- to 11-year olds. Instead, most providers screened based on family history of CVD or obesity. Most surprisingly, 62% and 89% believe that statins are appropriate for children and adolescents with LDL levels refractory to lifestyle modification but only 8% and 21% initiated statins (65).

 

Barriers to screening include health insurance availability and having a health care provider. Neither child’s age, family financial status, gender, obesity status, nor other health outcomes seemed to affect the likelihood of participating in lipid screening (41). Parents appeared to find lipid screening acceptable (66). However, in previous cascade screening programs of life-threatening FH where an index case leads to screening of 1st degree relatives, the prevalence of FH detected did not increase perhaps due to over 90% parents wanting possibly affected children to be screened but over 90% also wanting child testing to be done in the home (27,54,67,68). This preference has implications for lipid management logistics as well as inferences for parent preferences regarding minor children. Parental survey results in general population African-American families found most mothers of older children were in favor of cholesterol screening, but the majority of children with abnormal lipid levels did not return for follow-up due to doubts about test accuracy and the child’s anxiety or discomfort (69). Exacerbating the complicated parental attitudes are conflicted provider attitudes. Roughly three out of four providers believed future CVD risk could be prevented through pediatric lipid screening and treatment. But large majorities expressed lack of familiarity with pediatric lipid management while at the same time less than one-quarter would refer children to pediatric lipid specialists (70,71). So, lipid testing appears to be widely acceptable to families and providers, but with complex barriers to implementation.

EFFECTIVENESS AND EFFECTS OF SELECTIVE SCREENING

 

Since lipid screening in children appears to satisfy all WHO criteria for screening, it would be useful to know the benefits of screening. Following on the results of the CARDIAC study, recent data details the era of selective screening up to the NHLBI guidelines of late 2011 (72-74). The first such study in the modern era showed lipid testing rates in a geographically dispersed managed health care system network from 2002 to 2012 actually appear to have decreased (72). The proportion detected with severe FH-level LDL elevation did increase over time, but the yearly detection rate and cumulative incidence of those identified were far below the expected proportion of the cohort with FH. Within those tested each year, the proportion detected with moderately high LDL elevation or low HDL increased 5- to 9-fold at a time when nationally representative general pediatric population data indicated HDL levels had generally risen and LDL levels declined. Increased detection of low HDL-C and declining cohort mean HDL-C level led to an inference that providers were selectively testing youth with higher risk of having lifestyle dyslipidemia. Among those tested, the proportion with FH-level LDL was more than double the classic prevalence of FH suggesting that providers may also have been selectively screening youth with high risk of genetic dyslipidemia. A separate study based on 3 other managed care populations showed roughly similar screening proportions over a 3-year frame from 2007 to 2010, when accounting for cohort exclusions (74). In contrast, a study from the National Ambulatory Medical Care Survey (NAMCS) database showed an increasing trend in lipid testing, but with rates substantially lower than rates overall (73). The discrepancy may be related to NAMCS being composed of a national probability sample of physician self-reported data of outpatient encounters over a 1-week period. Using this approach, NAMCS data under-reports lipid testing by roughly 50% in adult patients (75). Moreover, the pediatric report included testing at well-child visits only and not subspecialty visits where high-risk youth may be more likely to be tested and treated (76).

 

Several studies have looked at screening after the promulgation of the 2011 NHLBI guidelines. Overall screening rates remain low but one study of patients in an ambulatory pediatric clinic demonstrated an increase in screening rates after 2011 from 17.1% to 20.1% (77). Other similar studies demonstrate no difference in screening patterns (78). Another study reviewed records from two pediatric clinics demonstrating only 27% adherence to the universal screening guidelines (79). With dismal screening rates many centers have implemented quality improvement efforts to increase screening rates. Peterson et al retrospectively reviewed charts of a general pediatric practice before and after guideline implementation, education initiatives, and EHR alerts demonstrating an increase in screening prevalence from 8.9% to 50% at the end of the study period (80). In a similar retrospective chart review an EHR prompt was created which required physicians to choose which lipid screening test was ordered or document why lipid screening was not performed. Lipid testing was also built into the 9, 10 and 11 year well child check order sets. With these efforts along with monthly data presentations by the QI team, authors showed a 64% increase in screening (81). In an alternate approach, a feasibility study on child-parent screening suggests testing at a well-child visit, particularly one where immunizations will be administered, as parents are primed for disease prevention (82).

 

Lipid screening does not necessarily lead to optimal outcomes. As noted, previous European data suggests a cascade screening approach did not substantively increase the prevalence of detected FH. In the CARDIAC universal screening study, parent telephone interviews were conducted between four and six weeks after screening. Only 40% of 342 respondents with at-risk children had made changes to their children’s diets in the immediate follow-up period and only 34% had modified physical activity (66). Data from the managed care network study showed that despite increased detection of severe dyslipidemia pharmacotherapy had not increased at all (72). The yearly rate of newly detected FH level LDL dwarfed the rate of pharmacotherapy initiation, signaling that screening for lipid abnormalities is not a panacea for improved lipid management. Finally, the International Childhood Cardiovascular Cohort Consortium found that incorporating lipid screening and clinical risk factor assessment provided a statistically significant improvement in prediction of cIMT in adulthood (83).

CONCLUSIONS AND FUTURE DIRECTIONS

 

Pediatric lipid testing appears to satisfy multiple criteria to make it worthy of wide screening. It is acceptable, accurate, repeatable, and testing is widely available. The natural history is well understood and childhood is a clear period of mounting severity but still latent and modifiable through acceptable therapies including lifestyle modification and simple pharmacotherapy. Accumulated data suggests selective screening is ineffective at detecting relevant cases and in translation to robust therapies lending support for universal screening programs. But several aspects are worthy of attention and future study in pursuing lipid testing of youth. Several aspects bolstering the success of universal screening efforts have been enumerated by the CARDIAC study investigators. Informational materials managing expectations about what happens on screening day, the risk factors assessed in the program, and follow up after the screening is useful. Another paramount task is effectively processing screening results and facilitating referral to treatment facilities, which requires cooperation between local hospitals, laboratories, and testing site. Testing programs can be leveraged to discuss primordial prevention and primary prevention in testing site or other locations where relevant children and families are gathered. Lipid results can also be integrated into broader health screening report that includes not only other assessment results but also broadly applicable treatment recommendations. Integrated these previously documented features into ongoing or future programs would be of great utility. Moving forward additional data needs to be gathered on the broader effects of universal screening. These effects may include dyslipidemia cases detected, the referral to lifestyle modification practitioners, pharmacotherapy initiation, and effects of therapy on improving lipid levels. Longer term studies are needed to document the CVD event risk modification stemming from early life CVD risk factor modification. Determinants of lipid testing, dyslipidemia identification, and lipid therapy need to be determined including at the patient, family, provider, practice, and geographic levels. While it appears to be a worthwhile endeavor, more study is urgently needed on improving the implementation of pediatric lipid screening.

 

REFERENCES

 

  1. Ford ES, Ajani UA, Croft JB, Critchley JA, Labarthe DR, Kottke TE, et al. Explaining the decrease in U.S. deaths from coronary disease, 1980-2000. N Engl J Med. 2007;356(23):2388-98.
  2. SPRINT Research Group, Wright JT, Jr., Williamson JD, Whelton PK, Snyder JK, Sink KM, et al. A Randomized Trial of Intensive versus Standard Blood-Pressure Control. N Engl J Med. 2015 Nov 26;373(22):2103-16. PubMed PMID: 26551272. Pubmed Central PMCID: 4689591.
  3. Ridker PM, Danielson E, Fonseca FA, Genest J, Gotto AM, Jr., Kastelein JJ, et al. Rosuvastatin to prevent vascular events in men and women with elevated C-reactive protein. N Engl J Med. 2008 Nov 20;359(21):2195-207. PubMed PMID: 18997196.
  4. Woolf SH, Schoomaker H. Life Expectancy and Mortality Rates in the United States, 1959-2017. JAMA.2019;322(20):1996–2016.
  5. Perak AM, Ning H, Kit BK, de Ferranti SD, Van Horn LV, Wilkins JT, & Lloyd-Jones DM (2019). Trends in Levels of Lipids and Apolipoprotein B in US Youths Aged 6 to 19 Years, 1999-2016. Jama, 321(19), 1895–1905.
  6. Expert panel on integrated guidelines for cardiovascular health and risk reduction in children and adolescents: summary report. Pediatrics. 2011;128 Suppl 5:S213-S56.
  7. American Academy of Pediatrics. National Cholesterol Education Program: Report of the Expert Panel on Blood Cholesterol Levels in Children and Adolescents. Pediatrics. 1992 Mar;89(3 Pt 2):525-84. PubMed PMID: 1538956. Epub 1992/03/01. eng.
  8. Services DoHaH. Patient Protection and Affordable Care Act. 2010 [cited 2015 January 2015]. Available from: http://www.gpo.gov/fdsys/pkg/BILLS-111hr3590enr/pdf/BILLS-111hr3590enr.pdf.
  9. de Ferranti SD, Steinberger J, Ameduri R, Baker A, Gooding H, Kelly AS, et al. (2019). Cardiovascular Risk Reduction in High-Risk Pediatric Patients: A Scientific Statement From the American Heart Association. Circulation, 139(13), 487–32.
  10. Newborn screening expands: recommendations for pediatricians and medical homes--implications for the system. Pediatrics. 2008;121(1):192-217.
  11. Wilson JMG, Jungner G. PRINCIPLES AND PRACTICE OF SCREENING FOR DISEASE. In: Organization WH, editor. Geneva: World Health Organization; 1968.
  12. Astrup A, Dyerberg J, Elwood P, Hermansen K, Hu FB, Jakobsen MU, et al. The role of reducing intakes of saturated fat in the prevention of cardiovascular disease: where does the evidence stand in 2010? Am J Clin Nutr. 2011 Apr;93(4):684-8. PubMed PMID: 21270379. Pubmed Central PMCID: 3138219. Epub 2011/01/29. eng.
  13. Brown MS, Goldstein JL. Familial hypercholesterolemia: A genetic defect in the low-density lipoprotein receptor. N Engl J Med. 1976;294(25):1386-90.
  14. Law MR, Wald NJ, Rudnicka AR. Quantifying effect of statins on low density lipoprotein cholesterol, ischaemic heart disease, and stroke: systematic review and meta-analysis. BMJ. 2003;326(7404):1423.
  15. O'Keefe JH, Jr., Cordain L, Harris WH, Moe RM, Vogel R. Optimal low-density lipoprotein is 50 to 70 mg/dl: lower is better and physiologically normal. J Am Coll Cardiol. 2004 Jun 2;43(11):2142-6. PubMed PMID: 15172426. Epub 2004/06/03. eng.
  16. Sachdeva A, Cannon CP, Deedwania PC, Labresh KA, Smith SC, Jr., Dai D, et al. Lipid levels in patients hospitalized with coronary artery disease: an analysis of 136,905 hospitalizations in Get With The Guidelines. Am Heart J. 2009 Jan;157(1):111-7 e2. PubMed PMID: 19081406. Epub 2008/12/17. eng.
  17. Taylor F, Ward K, Moore TH, Burke M, Davey SG, Casas JP, et al. Statins for the primary prevention of cardiovascular disease. Cochrane Database SystRev. 2011 (1):CD004816.
  18. Keech A, Simes RJ, Barter P, Best J, Scott R, Taskinen MR, et al. Effects of long-term fenofibrate therapy on cardiovascular events in 9795 people with type 2 diabetes mellitus (the FIELD study): randomised controlled trial. Lancet. 2005 Nov 26;366(9500):1849-61. PubMed PMID: 16310551.
  19. Pare G, Anand SS. Mendelian randomisation, triglycerides, and CHD. Lancet. 2010 May 8;375(9726):1584-6. PubMed PMID: 20452504.
  20. Holmes MV, Asselbergs FW, Palmer TM, Drenos F, Lanktree MB, Nelson CP, et al. Mendelian randomization of blood lipids for coronary heart disease. European heart journal. 2015 Mar 1;36(9):539-50. PubMed PMID: 24474739. Pubmed Central PMCID: PMC4344957.
  21. Rader DJ, Hobbs HH. Disorders of Lipoprotein Metabolism. In: Longo DL, Fauci AS, Kasper DL, Hauser SL, Jameson JL, Loscalzo J, editors. Harrison's Principles of Internal Medicine. 18. New York: McGraw-Hill; 2012.
  22. Barter PJ, Caulfield M, Eriksson M, Grundy SM, Kastelein JJ, Komajda M, et al. Effects of torcetrapib in patients at high risk for coronary events. N Engl J Med. 2007 Nov 22;357(21):2109-22. PubMed PMID: 17984165.
  23. Cuchel M, Bruckert E, Ginsberg HN, Raal FJ, Santos RD, Hegele RA, et al. (2014). Homozygous familial hypercholesterolaemia: new insights and guidance for clinicians to improve detection and clinical management. A position paper from the Consensus Panel on Familial Hypercholesterolaemia of the European Atherosclerosis Society. European Heart Journal, 35(32), 2146–2157.
  24. Wiegman A, Gidding SS, Watts GF, Chapman MJ, Ginsberg HN, Cuchel M, et al. Familial hypercholesterolaemia in children and adolescents: gaining decades of life by optimizing detection and treatment. European heart journal. 2015 Sep 21;36(36):2425-37. PubMed PMID: 26009596. Pubmed Central PMCID: 4576143.
  25. Daniels SR, Gidding SS, de Ferranti SD. Pediatric aspects of familial hypercholesterolemias: recommendations from the National Lipid Association Expert Panel on Familial Hypercholesterolemia. J Clin Lipidol. 2011 Jun;5(3 Suppl):S30-7. PubMed PMID: 21600527. Epub 2011/05/27. eng.
  26. Hopkins PN, Toth PP, Ballantyne CM, Rader DJ. Familial hypercholesterolemias: prevalence, genetics, diagnosis and screening recommendations from the National Lipid Association Expert Panel on Familial Hypercholesterolemia. J Clin Lipidol. 2011 Jun;5(3 Suppl):S9-17. PubMed PMID: 21600530. Epub 2011/05/27. eng.
  27. Williams RR, Hasstedt SJ, Wilson DE, Ash KO, Yanowitz FF, Reiber GE, et al. Evidence that men with familial hypercholesterolemia can avoid early coronary death. An analysis of 77 gene carriers in four Utah pedigrees. JAMA. 1986;255(2):219-24.
  28. Kusters DM, Avis HJ, de Groot E, Wijburg FA, Kastelein JJ, Wiegman A, et al. Ten-year follow-up after initiation of statin therapy in children with familial hypercholesterolemia. JAMA. 2014 Sep 10;312(10):1055-7. PubMed PMID: 25203086. Epub 2014/09/10. eng.
  29. Kit BK, Carroll MD, Lacher DA, Sorlie PD, J.M. D, C.L. O. Trends in serum lipids among US youths aged 6 to 19 years,1988-2010. JAMA. 2012;307(3):591-600.
  30. Kit BK, Kuklina E, Carroll MD, Ostchega Y, Freedman DS, Ogden CL. Prevalence of and Trends in Dyslipidemia and Blood Pressure Among US Children and Adolescents, 1999-2012. JAMA Pediatr. 2015 Jan 19. PubMed PMID: 25599372. Epub 2015/01/20. Eng.
  31. Berenson GS, Srinivasan SR, Bao W, Newman WP, 3rd, Tracy RE, Wattigney WA. Association between multiple cardiovascular risk factors and atherosclerosis in children and young adults. The Bogalusa Heart Study. N Engl J Med. 1998 Jun 4;338(23):1650-6. PubMed PMID: 9614255.
  32. Newman WP, 3rd, Freedman DS, Voors AW, Gard PD, Srinivasan SR, Cresanta JL, et al. Relation of serum lipoprotein levels and systolic blood pressure to early atherosclerosis. The Bogalusa Heart Study. N Engl J Med. 1986 Jan 16;314(3):138-44. PubMed PMID: 3455748.
  33. Relationship of atherosclerosis in young men to serum lipoprotein cholesterol concentrations and smoking. A preliminary report from the Pathobiological Determinants of Atherosclerosis in Youth (PDAY) Research Group. JAMA. 1990 Dec 19;264(23):3018-24. PubMed PMID: 2243430.
  34. Juhola J, Magnussen CG, Viikari JS, Kahonen M, Hutri-Kahonen N, Jula A, et al. Tracking of serum lipid levels, blood pressure, and body mass index from childhood to adulthood: the Cardiovascular Risk in Young Finns Study. J Pediatr. 2011 Oct;159(4):584-90. PubMed PMID: 21514597.
  35. Raitakari OT, Juonala M, Kahonen M, Taittonen L, Laitinen T, Maki-Torkko N, et al. Cardiovascular risk factors in childhood and carotid artery intima-media thickness in adulthood: the Cardiovascular Risk in Young Finns Study. JAMA. 2003 Nov 5;290(17):2277-83. PubMed PMID: 14600186. Epub 2003/11/06.
  36. Avis HJ, Vissers MN, Stein EA, Wijburg FA, Trip MD, Kastelein JJ, et al. A systematic review and meta-analysis of statin therapy in children with familial hypercholesterolemia. ArteriosclerThrombVasc Biol. 2007;27(8):1803-10.
  37. Versmissen J, Oosterveer DM, Yazdanpanah M, Defesche JC, Basart DC, Liem AH, et al. Efficacy of statins in familial hypercholesterolaemia: a long term cohort study. BMJ. 2008;337:a2423.
  38. Daniels SR, Greer FR. Lipid screening and cardiovascular health in childhood. Pediatrics. 2008;122(1):198-208.
  39. Risk of fatal coronary heart disease in familial hypercholesterolaemia. Scientific Steering Committee on behalf of the Simon Broome Register Group. BMJ. 1991 Oct 12;303(6807):893-6. PubMed PMID: 1933004. Pubmed Central PMCID: 1671226. Epub 1991/10/12. eng.
  40. May AL, Kuklina EV, Yoon PW. Prevalence of cardiovascular disease risk factors among US adolescents, 1999-2008. Pediatrics. 2012 Jun;129(6):1035-41. PubMed PMID: 22614778. Epub 2012/05/23. eng.
  41. Ritchie SK, Murphy EC, Ice C, Cottrell LA, Minor V, Elliott E, et al. Universal versus targeted blood cholesterol screening among youth: The CARDIAC project. Pediatrics. 2010 Aug;126(2):260-5. PubMed PMID: 20624798.
  42. Hollman G, Olsson AG, Ek AC. Disease knowledge and adherence to treatment in patients with familial hypercholesterolemia. The Journal of cardiovascular nursing. 2006 Mar-Apr;21(2):103-8. PubMed PMID: 16601526.
  43. Kools S, Kennedy C, Engler M, Engler M. Pediatric hyperlipidemia: child and adolescent disease understandings and perceptions about dietary adherence. Journal for specialists in pediatric nursing: JSPN. 2008 Jul;13(3):168-79. PubMed PMID: 18638047.
  44. Mackie TI, Tse LL, de Ferranti SD, Ryan HR, Leslie LK. Treatment decision making for adolescents with familial hypercholesterolemia: Role of family history and past experiences. J Clin Lipidol. 2015 Jul-Aug;9(4):583-93 e1-3. PubMed PMID: 26228677.
  45. Wierzbicki A, Ratcliffe C. The 2007 Heart-UK Survey of Lipid Clinics and Clinical Practice in the UK. Atherosclerosis. 2008;199:234.
  46. Braamskamp, MJAM, Kastelein JJP, Kusters DM, Hutten BA, Wiegman A (2016). Statin Initiation During Childhood in Patients With Familial Hypercholesterolemia. Journal of the American College of Cardiology, 67(4), 455–456.
  47. Vuorio A, Kuoppala J, Kovanen PT, Humphries SE, Strandberg T, Tonstad S, et al. Statins for children with familial hypercholesterolemia. Cochrane Database SystRev. 2010 (7):CD006401.
  48. Braamskamp MJAM,Langslet G, McCrindle BW, Cassiman D, Francis GA, Gagné C, Gaudet D, Morrison KM, Wiegman A, Turner T, Kusters DM, Miller E, Raichlen JS, Wissmar J, Martin PD, Stein EA, Kastelein JJP (2015). Efficacy and safety of rosuvastatin therapy in children and adolescents with familial hypercholesterolemia: Results from the CHARON study. Journal of Clinical Lipidology, 9(6), 741–750.
  49. Ference BA, Yoo W, Alesh I, Mahajan N, Mirowska KK, Mewada A, et al. Effect of long-term exposure to lower low-density lipoprotein cholesterol beginning early in life on the risk of coronary heart disease: a Mendelian randomization analysis. J Am Coll Cardiol. 2012 Dec 25;60(25):2631-9. PubMed PMID: 23083789. Epub 2012/10/23. eng.
  50. Keys A, Menotti A, Aravanis C, Blackburn H, Djordevic BS, Buzina R, et al. The seven countries study: 2,289 deaths in 15 years. Prev Med. 1984 Mar;13(2):141-54. PubMed PMID: 6739443. Epub 1984/03/01. eng.
  51. Brunner FJ, Waldeyer C, Ojeda F, Salomaa V, Kee F, Sans S, et al. (2019). Application of non-HDL cholesterol for population-based cardiovascular risk stratification: results from the Multinational Cardiovascular Risk Consortium. Lancet (London, England), 394(10215), 2173–2183.
  52. Efficacy and safety of lowering dietary intake of fat and cholesterol in children with elevated low-density lipoprotein cholesterol. The Dietary Intervention Study in Children (DISC). The Writing Group for the DISC Collaborative Research Group. JAMA. 1995 May 10;273(18):1429-35. PubMed PMID: 7723156.
  53. Lapinleimu H, Viikari J, Jokinen E, Salo P, Routi T, Leino A, et al. Prospective randomised trial in 1062 infants of diet low in saturated fat and cholesterol. Lancet. 1995 Feb 25;345(8948):471-6. PubMed PMID: 7861873.
  54. Marks D, Thorogood M, Neil SM, Humphries SE, Neil HA. Cascade screening for familial hypercholesterolaemia: implications of a pilot study for national screening programmes. Journal of medical screening. 2006;13(3):156-9. PubMed PMID: 17007658.
  55. Berwick DM, Cretin S, Keeler E. Cholesterol, children, and heart disease: an analysis of alternatives. Pediatrics. 1981 Nov;68(5):721-30. PubMed PMID: 6796932. Epub 1981/11/01. eng.
  56. Epstein LH, Kuller LH, Wing RR, Valoski A, McCurley J. The effect of weight control on lipid changes in obese children. American journal of diseases of children. 1989 Apr;143(4):454-7. PubMed PMID: 2929526.
  57. Jacobson MS, Tomopoulos S, Williams CL, Arden MR, Deckelbaum RJ, Starc TJ. Normal growth in high-risk hyperlipidemic children and adolescents with dietary intervention. Prev Med. 1998 Nov-Dec;27(6):775-80. PubMed PMID: 9922057. Epub 1999/01/28. eng.
  58. Kirk S, Brehm B, Saelens BE, Woo JG, Kissel E, D'Alessio D, et al. Role of carbohydrate modification in weight management among obese children: a randomized clinical trial. J Pediatr. 2012 Aug;161(2):320-7 e1. PubMed PMID: 22381024. Pubmed Central PMCID: 3406261.
  59. Siegel RM, Rich W, Joseph EC, Linhardt J, Knight J, Khoury J, et al. A 6-month, office-based, low-carbohydrate diet intervention in obese teens. Clinical pediatrics. 2009 Sep;48(7):745-9. PubMed PMID: 19264718.
  60. Sondike SB, Copperman N, Jacobson MS. Effects of a low-carbohydrate diet on weight loss and cardiovascular risk factor in overweight adolescents. J Pediatr. 2003 Mar;142(3):253-8. PubMed PMID: 12640371.
  61. Pratt RE, Kavey RE, Quinzi D. Combined dyslipidemia in obese children: response to a focused lifestyle approach. J Clin Lipidol. 2014 Mar-Apr;8(2):181-6. PubMed PMID: 24636177.
  62. Newman TB, Pletcher MJ, Hulley SB. Overly aggressive new guidelines for lipid screening in children: evidence of a broken process. Pediatrics. 2012 Aug;130(2):349-52. PubMed PMID: 22826571. Epub 2012/07/25. eng.
  63. Gillman MW, Daniels SR. Is universal pediatric lipid screening justified? JAMA. 2012;307(3):259-60.
  64. Psaty BM, Rivara FP. Universal screening and drug treatment of dyslipidemia in children and adolescents. JAMA. 2012;307(3):257-8.
  65. de Ferranti SD,Rodday AM, Parsons SK, Cull WL, O'Connor KG, Daniels SR, Leslie LK (2017). Cholesterol Screening and Treatment Practices and Preferences: A Survey of United States Pediatricians. The Journal of Pediatrics, 185, 99–105.e2.
  66. Cottrell L, John C, Murphy E, Lilly CL, Ritchie SK, Elliott E, et al. Individual-, family-, community-, and policy-level impact of a school-based cardiovascular risk detection screening program for children in underserved, rural areas: the CARDIAC Project. J Obes. 2013;2013:732579. PubMed PMID: 23840946. Pubmed Central PMCID: PMC3687496.
  67. Hardcastle SJ, Legge E, Laundy CS, Egan SJ, French R, Watts GF, et al. Patients' perceptions and experiences of familial hypercholesterolemia, cascade genetic screening and treatment. International journal of behavioral medicine. 2015 Feb;22(1):92-100. PubMed PMID: 24585182.
  68. Morris JK, Wald DS, Wald NJ. The evaluation of cascade testing for familial hypercholesterolemia. Am J Med Genet A. 2012 Jan;158A(1):78-84. PubMed PMID: 22139944. Epub 2011/12/06. eng.
  69. Price JH, Casler SM. African-American mothers' perceptions of cholesterol and its effects on their children. J Natl Med Assoc. 1996 Mar;88(3):145-50. PubMed PMID: 8839029. Pubmed Central PMCID: 2608033. Epub 1996/03/01. eng.
  70. Kimm SY, Payne GH, Stylianou MP, Waclawiw MA, Lichtenstein C. National trends in the management of cardiovascular disease risk factors in children: second NHLBI survey of primary care physicians. Pediatrics. 1998 Nov;102(5):E50. PubMed PMID: 9794980. Epub 1998/10/31. eng.
  71. Dixon DB, Kornblum AP, Steffen LM, Zhou X, Steinberger J. Implementation of Lipid Screening Guidelines in Children by Primary Pediatric Providers. J Pediatr. 2013 Nov 16. PubMed PMID: 24252785. Epub 2013/11/21. Eng.
  72. Zachariah JP, McNeal CJ, Copeland LA, Fang-Hollingsworth Y, Stock EM, Sun F, et al. Temporal trends in lipid screening and therapy among youth from 2002 to 2012. J Clin Lipidol. 2015 Sep-Oct;9(5 Suppl):S77-87. PubMed PMID: 26343215. Pubmed Central PMCID: 4562073.
  73. Vinci SR, Rifas-Shiman SL, Cheng JK, Mannix RC, Gillman MW, de Ferranti SD. Cholesterol testing among children and adolescents during health visits. JAMA. 2014 May 7;311(17):1804-7. PubMed PMID: 24794376. Epub 2014/05/06. eng.
  74. Margolis KL, Greenspan LC, Trower NK, Daley MF, Daniels SR, Lo JC, et al. Lipid screening in children and adolescents in community practice: 2007 to 2010. Circulation Cardiovascular quality and outcomes. 2014 Sep;7(5):718-26. PubMed PMID: 25160839. Pubmed Central PMCID: 4167939. Epub 2014/08/28. eng.
  75. Gilchrist VJ, Stange KC, Flocke SA, McCord G, Bourguet CC. A comparison of the National Ambulatory Medical Care Survey (NAMCS) measurement approach with direct observation of outpatient visits. Med Care. 2004 Mar;42(3):276-80. PubMed PMID: 15076827. Epub 2004/04/13. eng.
  76. Gregory ST, McNeal CJ, Copeland LA. Rates of cholesterol screening of youth. JAMA. 2014 Oct 1;312(13):1353-4. PubMed PMID: 25268451.
  77. Wilson DP,Davis S, Matches S, Shah D, Leung-Pineda V, Mou M, Hamilton L, McNeal CJ, Bowman WP(2015). Universal cholesterol screening of children in community-based ambulatory pediatric clinics. Journal of Clinical Lipidology, 9(S), S88–S92.
  78. Mihalopoulos NL,Stipelman C, Hemond J, Brown LL, Young PC (2018). Universal Lipid Screening in 9- to 11-Year-Olds Before and After 2011 Guidelines. Academic Pediatrics, 18(2), 196–199.
  79. Valle CW, Binns HJ, Quadri-Sheriff M, Benuck I, Patel A (2015). Physicians’ Lack of Adherence to National Heart, Lung, and Blood Institute Guidelines for Pediatric Lipid Screening. Clinical Pediatrics, 54(12), 1200–1205.
  80. DeSantes K,Dodge A, Eickhoff J, Peterson AL (2017). Improving Universal Pediatric Lipid Screening. The Journal of Pediatrics, 188, 87–90.
  81. Kern L, Crow J, Williams CB, Boies E, Gahagan S, Rhee KE (2017). Increasing Universal Lipid Screening Among 9- to 11-Year-Old Children Through a Quality Improvement Initiative. Clinical Pediatrics, 56(7), 640–647.
  82. Wald DS, Bestwick JP, Morris JK, Whyte K, Jenkins L, Wald NJ (2016). Child–Parent Familial Hypercholesterolemia Screening in Primary Care. New England Journal of Medicine, 375(17), 1628–1637.
  83. Koskinen J, Juonala M, Dwyer T, Venn A, Thomson R, Bazzano L, et al. (2018). Impact of Lipid Measurements in Youth in Addition to Conventional Clinic-Based Risk Factors on Predicting Preclinical Atherosclerosis in Adulthood. Circulation, 137(12), 1246–1255.

Genetic Etiology of Congenital Hypopituitarism

ABSTRACT

 

Congenital hypopituitarism refers to a deficiency of one or more pituitary hormones resulting from issues in fetal development. Embryological pituitary development involves a complex interplay of transcription factors, extrinsic and intrinsic to the oral ectoderm and neuroectoderm which develop to form the mature pituitary during early embryogenesis. Disruption of this process can result in isolated pituitary dysfunction, or effect nearby structures, such as the eye, olfactory bulbs, midline structures, and forebrain. Genetic causes make up an important portion of cases and have varying phenotypes even within identical mutations. This chapter provides an overview of pituitary development, and various causes of hypopituitarism including septo-optic dysplasia, syndromic and non-syndromic causes, isolated pituitary deficiencies, and syndromes associated with hypopituitarism. It also provides guidance on the investigation of genetic causes of hypopituitarism in the current genetic landscape.

 

INTRODUCTION

 

The pituitary gland can be thought of as the “hormonal control center” from which most endocrine organs are regulated. It is located in the sella turcica, posterior to the sphenoid sinus, inferior to the optic chiasm and the hypothalamus. The pituitary comprises 3 lobes, the anterior, intermediate and posterior.

 

The posterior lobe or neurohypophysis contains antidiuretic hormone (ADH or AVP) and oxytocin, contained in vesicles, as part of axonal projections from hypothalamic cells, known as the hypothalamic-hypophyseal tract. The intermediate lobe contains melanotrophs which produce alpha-melanocyte stimulating hormone (αMSH), although in humans is typically an embryological remnant and may not be present.

 

The anterior lobe or adenohypophysis contains 5 cell lines, producing 6 different hormones regulated by hypothalamic hormones via the hypophyseal portal system. Somatotrophs produce growth hormone (GH) while lactotrophs produce prolactin making up the somatomammotroph class of hormones. The glycoprotein hormone class consists of hormones with identical alpha subunits and different beta subunits, with thyroid stimulating hormone (TSH) produced by thyrotroph cells, and gonadotroph cells producing follicle stimulating hormone (FSH) and luteinizing hormone (LH). Corticotroph cells produce adrenocorticotrophic hormone (ACTH).

 

Growth hormone mediates its effect through induction of insulin like growth factor 1 (IGF-1) from the liver and is critical for linear growth in the child. Prolactin stimulates development of the mammary gland and lactation and is typically quiescent outside of pregnancy or childrearing. TSH stimulates production of thyroid hormone from the thyroid gland, which regulates metabolic rate and is critical for growth and cognitive development in early childhood. FSH and LH support gonadal development, production of testosterone or estrogen, and maturation of gamete cells. ACTH stimulates production of cortisol which is an essential stress hormone and ADH regulates plasma osmolality by inducing resorption of water in the collecting duct of the kidney.

 

Congenital hypopituitarism refers to a deficiency of one or more pituitary hormones resulting from events during fetal development. This may be the result of genetic mutation, antenatal insult, or as is commonly the case, be idiopathic. Pituitary deficiencies may be detected during the neonatal period; however, in some individuals’ pituitary deficiencies may not manifest until later in childhood. Furthermore, the severity can be variable.

 

Hypopituitarism may be isolated, affecting one cell line (isolated hormone deficiency), or multiple, termed combined or multiple pituitary hormone deficiency (CPHD, MPHD). Pan-hypopituitarism is typically used to describe deficiency of all anterior pituitary hormones; however, in some cases this term may also include the presence of AVP deficiency. AVP (or ADH) deficiency is termed as central diabetes insipidus (DI) but is undergoing a formal process to change the name to AVP deficiency. The new name is preferred as it highlights both etiology and treatment, while avoiding confusion with diabetes mellitus. AVP deficiency may coexist with other deficiency’s or be isolated.

 

The incidence of isolated growth hormone deficiency (IGHD) is reportedly 1 in 4000 live births (3), with CPHD also seen in 1 in 4000 live births (4), thus presenting an uncommon but important chronic condition.

 

Embryological pituitary development involves a complex interplay of transcription factors, extrinsic and intrinsic to the oral ectoderm and neuroectoderm which develop to form the mature pituitary. Disorders occurring during early development may affect nearby structures commonly the eye, olfactory bulbs, midline structures, and forebrain, whereas later events are typically isolated to pituitary abnormalities. Transcription factor gene defects have been demonstrated to cause different biochemical and structural forms of congenital hypopituitarism, although the majority of cases remain idiopathic. There is considerable phenotypic variability within known genetic causes of hypopituitarism, with some forms having incomplete penetrance, and presentation ranging from asymptomatic, to severe neonatal onset forms. This review discusses the clinical phenotypes of various genetic anomalies associated with hypopituitarism. Clinical manifestations and investigation of hypopituitarism are discussed in the “Hypopituitarism” chapter of Endotext.

 

This chapter will present an overview of the embryology of pituitary development before discussing the genetic causes of hypopituitarism. It will firstly discuss genes that cause septo-optic dysplasia, before discussing genes that cause other clinical phenotypes and hypopituitarism. After describing the genes associated with non-syndromic hypopituitarism, and isolated hormone deficiencies we will discuss other syndromes where hypopituitarism may be part of the phenotype. Each gene is described within the subheading that they most typically present; however, due to phenotypic variability some genes can present in multiple categories. For example, PROKR2 mutation classically causes isolated hypogonadotrophic hypogonadism, but can result in non-syndromic CPHD or septo-optic dysplasia in some cases (5-8). The information within this review is up to date as of May 2022; however, understanding of these genes continues to progress.

Figure 1. Stages of rodent pituitary development. (a) Oral ectoderm. (b) Rudimentary pouch. (c) Definitive pouch. (d) Adult pituitary gland. I infundibulum; NP neural plate; Nnotochord; PP pituitary placode; OM oral membrane; H heart; F forebrain; MB midbrain; HB hindbrain; RP Rathke's pouch; AN anterior neural pore; O oral cavity; PL posterior lobe; OC optic chiasm; P pontine flexure; PO pons; IL intermediate lobe; AL anterior lobe; DI diencephalon; SC sphenoid cartilage. Adapted from Sheng and Westphal, Trends in Genetics 1999;15:236-240, with permission (2).

EMBRYOLOGY

 

Pituitary gland development is classically expressed based on timing with murine development. The pituitary gland forms from the hypophyseal placode on the anterior neural ridge of the neural plate (evident embryonic (E) day 7.5 in the mouse model). Ventral displacement leads to the formation of the oral ectoderm and the roof of the mouth. A portion of this ectoderm thickens (E8.5) before invaginating rostrally at E9. At E9.5, corresponding with 4 weeks gestation, this migrates dorsally resulting in a cone shape, known as the rudimentary Rathke’s pouch. This continues to thicken with increased rostral mitosis spreading into the mesenchyme. Following ongoing proliferation, it separates from the oral ectoderm at E12.5 (approximately 6 weeks gestation). Hormonal cell progenitors arise from ventral proliferation forming the anterior pituitary. (Figure 1) The somatotrophs arise caudomedially, gonadotrophs rostroventrally and corticotrophs ventrally. The dorsal aspect later adjoins the descending infundibulum and remains thin (Intermediate lobe) (9).

 

Throughout this process the tissue is in contact with the neural plate neuroectoderm, at the base of the developing diencephalon. This neuroectoderm evaginates and progress dorsally to form the posterior pituitary. The tissue immediately ventral to this evagination forms the optic chiasm. The diencephalon develops throughout this process to form the hypothalamus (9).

 

The olfactory placode forms immediately lateral to the hypophyseal placode on the neural plate. These cells form the olfactory bulb but also the GnRH secreting cells which migrate to the forebrain from 6 weeks gestation, progressing to the hypothalamus by week 15. Axons from these cells extend into the hypothalamic portal system allowing regulation of gonadotroph cells (10-12). The optic nerve origin and optic chiasm develop from the neuroectoderm immediately anterior to the posterior pituitary, which has a complex interplay with some causes of hypopituitarism (13).

 

Correlation of pituitary development in mice to humans is difficult. Whilst pluripotent stem cells have been induced to form hypothalamic and anterior pituitary cells, the complex interplay of transcription factors, as well as interaction from other anatomical structures, mean these methods cannot give us a clear timeline in humans (14). Historical embryo samples demonstrate the formation of Rathke’s pouch by week 4, with interruption of connection to the oral cavity by week 6 with the anterior pituitary fully differentiated by 16 weeks (15).

 

Using hormone expression to correlate these two embryonic timelines, humans start producing GH, FSH, LH and ACTH at 8 weeks gestation, with TSH and prolactin at 13 weeks (9). Somatotrophs make up 50% of the cellular composition of the anterior pituitary, with 15-20% of cells being lactotrophs, 5% thyrotropes, 10-15% gonadotrophs and 15-20% corticotrophs (16) (Figure 2).

Figure 2. Structural development of pituitary. Lateral representation of process of oral and neuroectoderm folding. Cross-sectional representation of hormone producing pituitary cell groups. Adapted from Larkin S. et al, Endotext (1).

Various transcription factors are involved in coordinating these processes and gene mutation can result in underdevelopment or arrest of pituitary development. Figure 3 summarizes the temporal expression of various transcription factors within the pituitary. Abnormalities in genes expressed early in development such as SOX2, HESX1 and GLI2 more frequently effect other nearby structures, whereas those expressed later such as PROP1 and POU1F1 typically have localized effects. Transcription factors may be induced, upregulated or downregulated by multiple other transcription factors and the degree and site of expression can vary across the embryological development. For a diagrammatic overview of the role each transcription factor plays in pituitary development please see figure 1 of the 2020 JCEM review by Gregory L and Dattani M (4).

Figure 3. Timing of gene expression of transcription factors implicated in combined hypopituitarism.

Disruptions to this cascade of transcription factors causes a range of structural phenotypes, varying from a normal pituitary appearance, to anterior pituitary hypoplasia or agenesis. The infundibulum/pituitary stalk may be thin or absent and the posterior pituitary may be hypoplastic, absent, or ectopic. The combination of anterior pituitary hypoplasia (APH), infundibular thinning/absence, and ectopic posterior pituitary (EPP), is known as pituitary stalk interruption syndrome (PSIS) (Figure 4).

Figure 4. Lateral appearance of structural pituitary phenotypes. Adapted from Larkin et al, Endotext (1).

SEPTO-OPTIC DYSPLASIA

 

Septo-optic dysplasia (SOD) is defined by the presence of two or more of: optic nerve hypoplasia, hypopituitarism, and midline defects (agenesis of the corpus callosum or septum pellucidum) forming a clinical triad. SOD is a rare condition with incidence estimates between 1 in 9,000 to 1 in 40,000 (17,18). The association between hypopituitarism and optic nerve hypoplasia relates to the embryological origin of the optic chiasm being immediately anterior to the infundibular tissue, thus an insult at this location can interrupt both pathways. The phenotype is highly variable even in familial SOD (13). Although several genetic causes have been identified, these make up less than 10% of cases of SOD, and are summarized in Table 1 (19). Septo-optic dysplasia is commonly seen in young primiparous mothers (20) and while antenatal exposure to alcohol and drug use have been suggested, there is a lack of evidence to support causality (21).

 

Optic nerve hypoplasia (ONH) is typically bilateral (80-90%) with midline defects typically being septum pellucidum (85%) or corpus callosum (30%) hypoplasia or aplasia (21). 55-80% of cases have pituitary hormone deficiency, with growth hormone (GHD) and TSH deficiency (TSHD) being the most common deficiencies, followed by ACTH. Diabetes insipidus and hypogonadotrophic hypogonadism (HH) are found in less than 30% of hormone deficient cases (22-25). Approximately half of these hormone deficiencies are detected in the first 2 years of life and can present with severe neonatal hypoglycemia and cardiovascular instability, with the remainder of cases presenting across childhood to adolescence. The degree of deficiency is also noted to progress over time (22). Children with SOD typically have at least some degree of visual impairment, with 44- 80% being legally blind (22,26), while epilepsy and behavioral issues can be seen in approximately a quarter of patients, and cognitive impairment in 42.5% (22). Recently, 31% of cases were found to be obese despite therapy, suggesting the contribution of hypothalamic obesity (22).

 

HESX1

 

HESX1 is a member of the paired-like class of homeobox genes, located on chromosome 3p14.3 with autosomal dominant and recessive mutations described, making up approximately 1% of septo-optic dysplasia cases (19,27).

 

It appears from E8.5 through to E13.5 in the ventral diencephalon and oral ectoderm (28). HESX1 regulates the structural organization of the pituitary and interacts with the infundibulum and surrounding tissues through repressing transcription. Murine studies suggest absence typically does not prevent Rathke’s pouch/anterior pituitary formation (5%) but can result in polypituitary. It is suggested HESX1 regulates the regional expression of LHX3, PROP1, FGF8 and FGF10, preventing excessive expansion of the pituitary (19). HESX1 is upregulated by OTX2 and LHX3 transcription factors while being repressed by FGF8 and PROP1 (28,29).

 

Dattani et al. described a sibling pair with consanguineous parents, with a homozygous (R160C) HESX1 mutation in the late 1990’s. The SOD phenotype comprised optic nerve hypoplasia, midline defects (absent corpus callosum (CC) and septum pellucidum (SP)), ectopic posterior pituitary and anterior pituitary hypoplasia with hypopituitarism (30). Subsequent reports indicate that HESX1 mutation results in the complete SOD triad in approximately 30% of cases with ophthalmic changes including anophthalmia, microphthalmia, optic nerve hypoplasia and optic nerve aplasia (28,31). The majority of HESX1 pathogenic variants manifest GHD, with variability in deficiency of the other anterior hormones and ADH deficiency being uncommon. MRI appearance varies from normal to posterior pituitary ectopia and anterior pituitary hypoplasia in affected individuals (19). It is also associated with hypoplastic nasal cavities/olfactory bulbs and underdevelopment of the forebrain (28).

 

Autosomal dominant forms typically cause a milder phenotype, with undescended posterior pituitary and GHD, but phenotype-genotype correlation is poor (19). Penetrance is variable with asymptomatic parents and siblings being reported and midline defects and optic nerve anomalies absent in some cases (31,32). Testing of 217 patients with Kallmann syndrome (hypogonadotrophic hypogonadism (HH) and anosmia) revealed 1.4% had a mutation of HESX1 and normal pituitary on imaging (33).

 

SOX2

 

SOX2 and SOX3 are from the SOX (SRY-related high mobility group HMG Box) family of transcription factors and are both single exon genes that are expressed early in development and decline as cells differentiate. SOX 2 is located on chromosome 3q26.33 with autosomal dominant expression and cause 10-15% of microphthalmia and anophthalmia cases (19,34).

 

SOX2 is expressed during the morula stage, E2.5 but from gastrulation is restricted to the presumptive neuroectoderm, and E9.5 is found in the brain, CNS, sensory placodes, in the branchial arches, esophagus and trachea (28). At E11.5 it is expressed throughout Rathke’s pouch but from E18.5 is restricted to the intermediate lobe(35). In humans it is expressed within the developing anterior pituitary from weeks 3.5-9 and in the diencephalon but not in the neurohypophysis or infundibulum (19).

 

In 2006 Williamson et al. described heterozygous de novo mutations of SOX2 causing bilateral anophthalmia or severe microphthalmia associated with developmental delay, esophageal atresia and genital anomalies (36). Later that year, Kelberman et al. reported the association of SOX2 with HH, GHD and anterior pituitary hypoplasia (37). A cohort of 18 participants with SOX2 mutation showed 33% (6/18) had short stature or GHD, and 44% (8/18) had genital anomalies or hypogonadism (38).

 

Alongside hypoplastic anterior pituitary, hypothalamic hamartomas and corpus callosum abnormalities are often seen, as are hippocampus anomalies, sensorineural hearing loss, learning difficulties and cognitive impairment. The vast majority have anophthalmia in at least one eye, but milder cases have microphthalmia, coloboma and optic nerve hypoplasia, with normal ocular structures seen very rarely (34). SOX2 mutations are associated with esophageal/tracheal abnormalities and underdeveloped genitalia known as Anophthalmia-Esophageal-Genital (AEG) syndrome. Micropenis and cryptorchidism are commonly seen, and while genital anomalies are noted less commonly in females, they can be severe, with some cases having vaginal agenesis (28).

 

Anophthalmia or microphthalmia with esophageal abnormalities, sensorineural hearing loss or GHD and HH should raise the question of a SOX2 mutation for a clinician. Given the autosomal dominant inheritance, detection would be critical for family planning for both the patient and their parents.

 

SOX3

 

SOX3 gene is found at Xq27 causing X-linked recessive inheritance found to cause 0.2% of all GH deficient cases (28,39). SOX3 is expressed throughout the central nervous system during embryonic development.

 

It is first expressed at E6.5 but is rapidly restricted to the anterior ectoderm, and to the presumptive neuroectoderm. It is highly expressed in the infundibulum and presumptive hypothalamus but not in Rathke’s pouch (28).

 

SOX3 supports development of the hypothalamus and infundibulum as well as the development of the AP. Absence results in thinning of the infundibulum, which is thought to reduce expression of other genes involved with supporting pituitary proliferation. As a result, Rathke’s pouch is initially expanded but eventually the anterior pituitary becomes hypoplastic. It is important in confining the expression of FGF8 and BMP4, preventing excessive activity. Interestingly, duplications of SOX3 are also associated with similar phenotypes to those with non-functional mutations (typically polyalanine expansions). This suggests that correct dose of SOX3 is required for optimal embryonic development (28).

 

In 2002, Laumonnier et al. described a boy with IGHD and learning difficulties associated with a SOX3 mutation (40). Subsequently, GHD has been found in affected males and is associated with developmental delay or cognitive impairment of varying severity. One family of 5 affected females with short stature and language/ hearing impairment has been described (28). Other concurrent pituitary hormone deficiencies are present with variable frequency with panhypopituitarism described in some cases (28).

 

MRI findings include absent corpus callosum and absent or hypoplastic infundibulum and ectopic posterior pituitary, with cognitive impairment and learning difficulties also reported. SOX3 is not associated with optic nerve hypoplasia and has not been reported in patients with the complete SOD triad. Craniofacial abnormalities were reported frequently in murine models but are rare in human populations and 1 case of spina bifida has been reported (28).

 

Patients with an x-linked pattern of inheritance, and isolated GHD or CPHD should raise clinical suspicion for SOX3 mutation.

 

OTX2

 

Orthodentic Homeobox 2 (OTX2) is critical for anterior structure development and forebrain maintenance (28). It is located on chromosome 14q22.3 with an autosomal dominant inheritance that has been detected in 2-3% of cases of anophthalmia/microphthalmia and can be associated with hypopituitarism (29,38,41).

 

From E10.5 OTX2 is strongly expressed in the neurohypophysis and hypothalamus but the expression in Rathke’s pouch is minimal. Expression continues until E16.5 but is silenced by E12.5. Knockout within Rathke’s pouch caused no abnormalities in pituitary development or function (42). OTX2 deficiency results in decreased posterior lobe and pituitary stalk development and secondary anterior lobe hypoplasia. This is mediated through failure of FGF10 production, resulting in the hypoplastic anterior pituitary, but with normal cell differentiation (42). It is also important for upregulating HESX1 and POU1F1 (29).

 

Isolated GHD is classical for OTX2 mutations but CPHD, typically TSHD or HH and panhypopituitarism have been described. Imaging typically demonstrates ectopic or absent posterior pituitary with hypoplastic or normal anterior pituitary but may be normal (28,41). Ocular manifestations vary from anophthalmia/microphthalmia, to optic nerve hypoplasia or retinal dystrophy, with normal ocular phenotype also described. Additional anomalies are rarely reported (41).

 

OTX2 abnormalities should be considered in cases of anophthalmia/microphthalmia and hypopituitarism. Posterior pituitary anomalies and absence of extracranial features may differentiate from SOX2 mutations (28,41).

 

PAX6

 

Other genes known to be associated with anophthalmia/microphthalmia have been shown to causes septo-optic dysplasia and hypopituitarism. PAX6 is located on chromosome 11p13 and is inherited in autosomal dominant pattern with incomplete penetrance (43). In murine pituitary development, PAX6 is expressed in the dorsal side of Rathke’s pouch from E9-12.5 and establishes the ventral-dorsal cell boundaries (44). It has a major role in early eye development, classically causing aniridia, with optic nerve anomalies including hypoplasia or microphthalmia or anophthalmia also described (43). Over 500 cases of PAX6 mutation have been described with only four cases of hypopituitarism, including isolated ACTH deficiency in one, GH deficiency in the remainder, two of which had plausible HH. Two were noted to have a hypoplastic pituitary (45-47). Thus, PAX6 is a rare cause of hypopituitarism in patients with eye abnormalities and may present with SOD.

 

RAX

 

The RAX gene found on chromosome 18q21.32 is involved in forebrain and eye development and is inherited in an autosomal recessive nature. In humans, it is associated with anophthalmia, microphthalmia and palatal anomalies and is present in approximately 3% of anophthalmia/microphthalmia cases (38). One patient to date with a severe mutation had anterior and posterior pituitary agenesis on MRI resulting in panhypopituitarism, but other patients have not been found to have pituitary anomalies (48).

 

TCF7L1

 

The transcription factor 7-like 1 (TCF7L1) gene on chromosome 2p11.2 is a transcription regulator gene, important for brain development through WNT/β-catenin signaling pathway. Heterozygous mutations of TCF7L1 have been identified in 2 SOD patients, 1 of whom had GHD and ACTHD and MRI demonstrating ONH, anterior pituitary hypoplasia and absent posterior pituitary (49).

 

Other Genes

 

GLI2, BMP4, TBC1D32, PROKR2, FGF8, FGFR1 mutations are also associated with SOD but are described later as this is not their typical phenotype. Genes causing SOD are summarized in Table 1.

.

Table 1. Genes Associated with Septo-Optic Dysplasia

Gene

Inheritance/ prevalence

Ocular Phenotype

Pituitary appearance

Pituitary deficiencies

Other Features

HESX1

Chr 3p14.3

AD/AR (incomplete penetrance)

1% of SOD

ONH

EPP, APH

GH, CPHD,

ACTH + ADH less common

CC/SP changes, hypoplastic olfactory, underdeveloped forebrain

SOX2

Chr 3q26.33

AD

10-15% of AO/MO

AO/MO

APH, hypothalamic hamartoma

GH, LH/FSH

CC changes, SNHL, ID,
esophageal anomalies, genital anomalies

SOX3

Chr Xq27

XR

0.4% of IGHD

Nil

Thin infundibulum, EPP, APH, PSIS

GH,

CPHD uncommon

CC changes, ID

OTX2

Chr 14q22.3

AD

3% of AO/MO

AO/MO, ONH, retinal dystrophy

EPP, APH, PSIS

GH,

LH/FSH, TSH.

ACTH less common

 

PAX6

Chr 11p13

AD

Rare

Aniridia, ONH, AO/MO

APH

Rare

GH,

ACTH, FSH/LH

ASD, ADHD, obesity, diabetes mellitus

RAX

18q21.32

AR

3% of AO/MO

AO/MO

 

Rare

CPHD

Palate changes

TCF7L1

Chr 2p11.2

AD

Rare

ONH

Normal, APH, absent posterior pituitary

GH, ACTH

CC/SP changes

GLI2

2q14.2

AD

1-13% of CPHD

ONH

APH, EPP, PSIS

GH, ACTH,

FSH/LH, TSH

SP/CC changes, polydactyly, midface hypoplasia, cleft palate/lip, HPE

BMP4

Chr 14q22-23

AD

Rare

AO/MO

Normal

GH, TSH

Myopia, cleft palate/lip, polydactyly

 

TBC1D32

Chr 6q22.31

AR

Rare

ONH

APH, EPP or absent pituitary

GH, CPHD

Oro-facial-digital syndrome, CC agenesis

PROKR2

Chr 20p12.3

AD/AR

5-23% of HH

ONH

PSIS

HH, CPHD

 

FGF8

Chr 10q24

AD/AR

1% of HH

ONH

 

HH, CPHD

HPE, VACTERL

FGFR1

Chr 8p11.2p12

AD

5-11% of HH

ONH

PSIS

HH, CPHD

Split hand/foot malformation

AD – Autosomal Dominant, ADHD – Attention Deficit Hyperactivity Disorder, AO – Anophthalmia, APH – Anterior Pituitary Hypoplasia, AR – Autosomal Recessive, ASD – Autism Spectrum Disorder, CC– Corpus Callosum, CPHD - Combined Pituitary Hormone Deficiency, EPP – Ectopic Posterior Pituitary, HH – Hypogonadotrophic Hypogonadism, HPE – Holoprosencephaly, ID – Intellectual Disability, MO – Microphthalmia, ONH – Optic Nerve Hypoplasia, PSIS – Pituitary Stalk Interruption Syndrome, SP – Septum Pellucidum, SNHL – Sensorineural Hearing Loss, VACTERL – Vertebral defects, Anal atresia, Cardiac defects, Tracheo-esophageal fistula, Renal anomalies, and Limb abnormalities.

 

SYNDROMIC CAUSES OF HYPOPITUITARISM

 

Septo-optic dysplasia is the most common syndromic presentation of hypopituitarism. Other genes causing hypopituitarism, which do not typically cause optic nerve hypoplasia are discussed below, although GLI2 and BMP4 may present with SOD. Syndromic causes of hypopituitarism are summarized in Table 3.

 

LHX3

 

The LIM homeobox proteins LHX3 and LHX4 are expressed in early development and have a co-dependent role in supporting pituitary development. The LHX3 gene located on chromosome 9q34.3 has autosomal recessive expression and is seen in 0.5-1.2% of hypopituitarism cases (39,50,51).

 

LHX3 is first expressed in Rathke’s pouch at E9.5 following induction by FGF8 and by E12.5 is predominantly expressed in the dorsal aspect of the pouch. LHX3 is also expressed in the ventral hindbrain, inner ear and spinal cord, and is upregulated by LHX4 in the pituitary and SOX2 in the inner ear. LHX3 is expressed in the anterior and intermediate pituitary throughout development, persisting into adulthood and is thus thought to have a role in maintaining some of the cell types. In LHX3 null mice, Rathke’s pouch is initially formed, but regresses resulting in aplasia of the anterior and intermediate lobes, which is at least in part due to failure to support HESX1 and POU1F1 production. LHX3 is critical for maturation of Rathke’s pouch into the anterior pituitary and regulates differentiation of all anterior cell lines, although corticotrophs are somewhat preserved (52).

 

As a result, LHX3 mutations commonly cause CPHD or panhypopituitarism, with ACTH production normal in approximately half. Children typically present with CPHD at birth but milder phenotypes of developing hypopituitarism throughout childhood are described. A variable appearance is seen on imaging, ranging from normal (10%) to aplasia or hyperplasia, with one case having a microadenoma type appearance (53). Sensorineural hearing loss is seen in up to 50% of cases as LHX3 is present in a restricted region of the inner ear in tandem with SOX2. Intellectual or learning difficulties are reported in 38% of cases (51). A short stiff neck with limited rotation is seen in approximately 70% of cases, with other vertebral anomalies such as scoliosis seen in approximately 50% of cases. Respiratory distress has occasionally been described (54,55).

 

LHX3 should be suspected in patients with early-onset CPHD with a stiff neck and/or hearing impairment. Neonatal respiratory distress is a rare but important association that may also point to LHX3 mutation.

 

LHX4

 

The LHX4 gene is found at chromosome 1q25.2 with autosomal dominant expression causing 0.9-1.4% of hypopituitarism cases (50,56).

 

LHX4 is expressed in Rathke’s pouch from E9.5 like LHX3; however, by E12.5 it is restricted to the anterior lobe. It also expressed in hindbrain, cerebral cortex and spinal cord tissues. LHX4 and LHX3 work in tandem to support specialization of pituitary cells and LHX4 is important for upregulating LHX3 and POU1F1 and ensuring correct timing of this induction. Thus, LHX4 deficiency may result in an element of LHX3 deficiency and causes a hypoplastic anterior pituitary with appropriate cell differentiation but reduced numbers of hormone producing cells (2,57-59).

 

Like in LHX3, LHX4 also typically results in CPHD, but milder presentations, including transient isolated GHD, have been reported (56). One family of recessive lethal LHX4 has been reported with severe combined hypopituitarism and fatal respiratory distress thought to be secondary to their hypopituitarism (60). Appearances are again variable on imaging; ectopic posterior pituitary is common with the anterior pituitary ranging from normal to aplasia. Chiari 1 malformation is also associated but likely represents a minority of cases (56). Cardiac defects have been reported in 2 cases and respiratory distress in 4 cases (56,60-62). Penetrance is variable with asymptomatic effected parents being a common occurrence (56).

 

LHX4 mutation should be considered in patients with Chiari malformation or respiratory/cardiac defects in a patient with hypopituitarism.

 

GLI2

 

The GLI2 gene is a zinc finger transcription factor found on chromosome 2q14.2 and has an autosomal dominant inheritance with incomplete penetrance (63). Whilst prevalence has been reported in as many of 13.6% of cases of hypopituitarism, other cohorts have found it to be rare (<1%) (39,63).

 

GLI2 is expressed in the ventral diencephalon, where it induces BMP4 and FGF8 and is present in the oral ectoderm and Rathke’s pouch from E8.5, where it is important for mediating SHH (Sonic Hedgehog) response. GLI2 is important for supporting pituitary progenitor proliferation and its absence results in reduced pituitary cell numbers, particularly corticotrophs, somatotrophs and lactotrophs, but appropriate differentiation of cells in general (64).

 

GLI2 has been shown to mediate SHH effect and causes of holoprosencephaly (HPE) are commonly known to affect SHH. In 2003, Roessler et al. identified GLI2 gene mutations in 7 out of 390 patients meeting HPE criteria (65); however, this has since been shown to be an uncommon manifestation GLI2 (66). A study of 112 cases with GLI2 mutations found only three children with holoprosencephaly, and one of whom had an alternative causative gene detected. This study showed that of those with a truncated GLI2 mutation, 62% had pituitary abnormalities and polydactyly and only 15% did not have either anomaly. 58% of those with non-truncated variants had hypopituitarism. 13% had midface hypoplasia with 16% having cleft palate/lip (66). Unlike other causes of holoprosencephaly, GLI2 typically manifests with anterior pituitary anomalies, particularly GH and ACTH deficiency (63).

 

MRI findings vary, including optic nerve hypoplasia, septum pellucidum or corpus callosum hypoplasia/agenesis, anterior pituitary hypoplasia, ectopic posterior pituitary or PSIS. Thus SOD, PSIS, and rarely HPE are all plausible manifestations of GLI2 mutation (67).

 

The presence of anterior pituitary deficiency and polydactyly or midface abnormalities/central incisor should raise suspicion for GLI2 and trigger further investigation.

 

GLI3

 

Heterozygous mutation of another zinc finger transcription factor GLI3 found on chromosome 7p14.1, results in Pallister-Hall syndrome which presents with mesoaxial polydactyly and hypothalamic hamartoma (68). In mouse studies, GLI3 is not critical for pituitary or hypothalamic development or differentiation, but if combined with GLI2 mutation, the phenotype is more severe than GLI2 anomalies alone (64,69). Mutation in the amino-terminal third of the GLI3 gene results in Greig Cephalosyndactyly syndrome, associated with polydactyly, macrocephaly and hypotelorism, without hypothalamic abnormalities, with Pallister Hall being caused by mutations in the middle third of the gene (70).

 

In 1980 Hall and Pallister et al. described 6 cases with hypothalamic hamartoma, panhypopituitarism, postaxial polydactyly and imperforate anus, with lethality in all cases (71). Since this time, milder/non-lethal familial forms have been denoted, with the pituitary phenotype varying from normal pituitary function to isolated GHD and panhypopituitarism, thus all cases should be assessed for pituitary dysfunction (72). As with other causes of hypothalamic hamartoma, central precocious puberty can also be associated and should be monitored for (68).

 

Hypopituitarism Associated With Holoprosencephaly

 

The prosencephalon cleaves into right and left hemispheres between day 18 and 28 gestation in humans. Failure of this process results in holoprosencephaly (HPE) of varying degrees with lobar holoprosencephaly having separated 3rd ventricles with a connection within the frontal cortex, semilobarholoprosencephaly being partially separated and alobarholoprosencephaly having a continuous single anterior ventricle. (Figure 5) Facial development is affected by the same process and can cause hypotelorism or cleft palate, but also severe cases may have anophthalmia or cyclopia with a superiorly displaced proboscis. Holoprosencephaly is present in 1 in 250 embryos’ but there is a high rate of fetal demise. It is estimated to occur in 1 in 8,000-16,000 live births (73).

Figure 5. Types of holoprosencephaly.

Other associated midline defects include underdevelopment of olfactory bulbs and corpus callosum, single midline maxillary incisor and hypoplastic pituitary or ectopic posterior pituitary. Holoprosencephaly is associated with anterior hypopituitarism in 5-10% of cases, with 70% having diabetes insipidus (DI) (74).

 

Many cases are caused by trisomy 13 making up 40-60% of all causes with a genetic anomaly, trisomy 18 and triploidy are also associated. Sonic hedgehog and ZIC2 mutations make up 5% of non-syndromic cases each. Importantly, structural chromosomal anomalies in almost all chromosomes have been shown to cause holoprosencephaly (75,76).

 

The SHH gene on chromosome 7q36 is critical for early development of the CNS, particularly the forebrain and is present in the notochord, neural tube and posterior limb buds of the gut. It is a major gene implicated in holoprosencephaly and its effect is mediated through other transcription factors, many of which have been demonstrated to cause HPE shown in Table 2 (73).

 

There is limited data regarding the rates of anterior pituitary dysfunction with each gene type although cases in SHH and ZIC2 have been reported (67,77).

 

Table 2. Genes Associated with Holoprosencephaly and Hypopituitarism

Gene

Chromosome

% of HPE

Other Features

SHH

7q36

5.4-5.9%

Renal-urinary anomalies

ZIC2

13q32

4.8-5.2%

Renal-urinary anomalies

SIX3

2p21

3%

Typically, severe HPE phenotype

TGIF

18p11.3

<1%

Wide spectrum of severity

CDON

11q24.2

rare

CHD, renal dysplasia, radial defects, gallbladder agenesis

CHD – Congenital Heart Disease, HPE – Holoprosencephaly. Adapted from Tekendo-Ngongang C, et al. Holoprosencephaly Overview. GeneReviews. 2020 (76).

 

Pituitary Stalk Interruption Syndrome

 

Pituitary stalk interruption syndrome (PSIS) is a triad of a thin/absent pituitary stalk, ectopic posterior pituitary and aplasia/hypoplasia of the anterior pituitary (Figure 6). SHH and other downstream genes of the SHH signaling pathway, including SIX3, TGIF, CDON and GPR161 have all been reported in 1-2 cases with PSIS and CPHD, without holoprosencephaly (76,78-82). Other causes of hypopituitarism that have been associated include POU1F1, PROP1, LHX3, LHX4, HESX1, SOX3, OTX2, PROKR2 and FOXA2. 100% of patients have been GHD in large case series with gonadotrophin, ACTH and TSH deficiencies in the majority and hyperprolactinemia in a minority (83).

Figure 6. Lateral appearance of pituitary stalk interruption syndrome (PSIS). Adapted from Larkin S. et al, Endotext (1).

ROBO1

 

Heterozygous ROBO1 gene mutations (found on chromosome 3p12.3) has been described in 5 patients with PSIS, with a 6th case of homozygous mutation (84,85). Ocular anomalies, including hypermetropia and ptosis were also seen, but penetrance was incomplete with unaffected parents in some cases. Combined GHD and TSHD, and a case of panhypopituitarism have been described (84).

 

FOXA2

 

The forkhead box A2 (FOXA2) gene is found on chromosome 20p11.21 with dominant or de novo inheritance (86). FOXA2 regulates expression of GLI2 and SHH and mutations of the FOXA2 gene and 20p11 deletions have been reported to cause hypopituitarism (87-89). Imaging demonstrates anterior pituitary hypoplasia, absent or ectopic posterior pituitary and absent or thin pituitary stalk (86,90). FOXA2 is also involved in regulation of the ABCC8 and KCNJ11 genes, critical for the K-ATP channels in pancreatic beta-cells, and mutation has been shown to cause congenital hyperinsulinism (91). Other phenotypic features include craniofacial dysmorphism, choroidal coloboma and malformations of the liver, lung and heart (90,92,93).

 

EIF2S3

 

Mutations in the EIF2S3 gene found on chromosome Xp22.11, result in the MEHMO syndrome (Mental retardation, Epileptic seizures, Hypogenitalism, Microcephaly and Obesity). EIF2S3 is expressed in the pituitary and hypothalamus during embryological development (94). Mutation typically results in GHD, and panhypopituitarism has been described (94,95). Imaging typically demonstrates corpus callosum thinning and either normal, or hypoplastic anterior pituitary (94).

 

BMP4

 

Bone morphogenetic protein 4 (BMP4) gene is found on chromosome 14q22-q23 with dominant inheritance and incomplete penetrance (70). BMP4 is expressed in the diencephalon and infundibulum from E8.5, and absence results in failure of pituitary placode and Rathke’s pouch development in mouse studies (96). Mutations are frequently associated with anophthalmia/microphthalmia, cleft lip/palate and polydactyly/syndactyly. In 2018 Rodríguez-Contreras et al. reported a pathogenic BMP4 mutation in a boy with GH and TSH deficiency and myopia, but further cases have not yet been described (97).

 

ARNT2

 

Chromosome 15q25.1 is the locus for the aryl hydrocarbon receptor nuclear translocator 2 (ARNT2) gene and has autosomal recessive inheritance. ARNT2 is expressed in the developing anterior and posterior pituitary and hypothalamus and mutations result in CPHD with diabetes insipidus. Pituitary imaging demonstrates absent posterior pituitary bright spot with stalk thinning and a hypoplastic anterior pituitary. It is also associated with postnatal microcephaly, hypoplastic frontal and temporal lobes, renal anomalies and vision impairment (98).

 

TBC1D32

 

The TBC1 domain family member 32 (TBC1D32) gene is found at Chromosome 6q22.31 and results in disease with an autosomal recessive inheritance. TBC2D32 is expressed in the developing hypothalamus and Rathke’s pouch and is involved in SHH signaling and ciliary functioning. Isolated GHD or panhypopituitarism were found in the limited cases reported to date. Anterior pituitary hypoplasia or aplasia is described with ectopic or absent posterior pituitary as is partial agenesis of the corpus callosum and optic chiasm hypoplasia. Other phenotypic features include oral anomalies (cleft palate), facial dysmorphism (hypertelorism/cleft lip) and limb anomalies (polydactyly/syndactyly), consistent with oro-facial-digital syndrome (99).

 

Table 3. Syndromic Causes of Hypopituitarism

Gene

Inheritance/ prevalence

SOD/

PSIS/

HPE

Pituitary appearance

Pituitary deficiencies

Other Features

LHX3

Chr 9q34.3

AR

0.5-1.2%

No

APH, Normal, hyperplasia

CPHD,
ACTH in 50%

SNHL, ID, short stiff neck, scoliosis, rarely RDS

LHX4

1q25.2

AD

0.9-1.4%

No

APH, EPP

CPHD

CC hypoplasia, Chiari 1 malformation, rarely CHD/RDS

GLI2

2q14.2

AD

1-13%

SOD, HPE, PSIS

APH, EPP, PSIS

GH, ACTH,

FSH/LH, TSH

ONH, SP/CC changes, polydactyly, midface hypoplasia, cleft palate/lip

GLI3

Chr 7p14.1

AD

Rare

No

Hypothalamic hamartoma

GH,

ACTH, FSH/LH, TSH

Postaxial polydactyly, imperforate anus. CPP

ROBO1

Chr 3p12.3

AD/AR

Rare

PSIS

PSIS

GH, TSH

Pan-hypopituitarism

Hypermetropia, ptosis

EIF2S3

Chr Xp22.11

XR

No

Normal, APH

GH,

Pan-hypopituitarism

CC hypoplasia, ID, epilepsy, gonadal failure, microcephaly, obesity

BMP4

Chr 14q22-23

AD

Rare

SOD

Normal

GH, TSH

AO/MO, myopia, cleft palate/lip, polydactyly

FOXA2

Chr 20p11.21

AD/De novo

PSIS

APH, EPP,

PSIS

Pan-hypopituitarism,

Hyperinsulinism, craniofacial dysmorphism, choroidal coloboma, and liver, lung and heart malformations

ARNT2

Chr 15q25.1

AR

No

APH, thin stalk, absent PP

Pan-hypopituitarism, ADH

Microcephaly, fronto-temporal hypoplasia, renal anomalies and vision impairment.

TBC1D32

Chr 6q22.31

AR

SOD

APH, EPP or absent pituitary

GH, pan-hypopituitarism

Oro-facial-digital syndrome, CC agenesis

HPE Genes  

-       SHH

-       ZIC2

-       SIX3

-       TGIF

-       CDON

Rare

HPE,

PSIS

Normal

Rarely PSIS

ADH

Rarely CPHD

 

             

AO – Anophthalmia, APH – Anterior Pituitary Hypoplasia, CC – Corpus Callosum, CHD – Congenital Heart Disease, CPP – Central Precocious Puberty, EPP – Ectopic Posterior Pituitary, FTT – Failure To Thrive, HPE – Holoprosencephaly, ID – Intellectual Disability, MO – Microphthalmia, PP – Posterior Pituitary, PSIS – Pituitary Stalk Interruption Syndrome, RDS - Respiratory Distress, SOD – Septo-optic Dysplasia, SNHL – sensorineural hearing loss, SP – Septum Pellucidum, XR – X-linked Recessive

 

NON-SYNDROMIC CAUSES OF HYPOPITUITARISM

 

POU1F1

Figure 7. Cross-sectional representation of pituitary cell groups in POU1F1. Adapted from Larkin S. et al, Endotext (1).

POU1F1 was the first genetic cause of CPHD detected, initially described in mice (known as PIT-1) and 2 years later in humans. It is a member of the POU family of transcription factors, and the gene is located at 3p11.2. Approximately 65% of cases are autosomal recessive (homozygous/compound heterozygous) with 35% having an autosomal dominant form, the most common being the R271W mutation. In a review of 15 cases, the majority had affected parents, with only 3 cases of hormonally intact parents (100). POU1F1 is present in 2.8% of CPHD, with up to 21% of familial CPHD (50). There is considerable variability between countries, with one cohort of 23 CPHD patients in India detected 6 POU1F1 mutations and many other studies having low numbers of detected mutations (50,100,101).

 

Expression of POU1F1 is triggered by PROP1 and is seen within the pituitary from E13.5, persisting through to adulthood. It is critical for supporting the differentiation and proliferation of thyrotroph, lactotroph and somatotroph cells, and stimulates the GH1, prolactin and TSHB genes, required for hormone synthesis (102). LHX3, LHX4 and OTX2 upregulate POU1F1 but PROP1 is essential for its function (52,57,59).

 

Tatsumi et al. detected the mutation in a case of “cretinism” with cognitive impairment and short stature, with GH, TSH and prolactin deficiency, reported in 1992 (103). 100% of cases in the literature have GH deficiency with 87.5% having TSH deficiency and 95.6% having prolactin deficiency (demonstrated in Figure 7). TSH deficiency is typically detected in infancy, either prior to, or simultaneously with GH and prolactin deficiency, with ~13.5% detected after GHD. 70% have anterior pituitary hypoplasia, but other midline and peripheral defects are rarely seen. Autosomal dominant forms are less likely to have an absent pituitary (63.6%) and typically have less severe GHD with 20% having an undetectable GH peak on stimulation testing, compared to 48%. Apart from hypopituitarism, there are no other phenotypic features known to be associated with POU1F1 (100).

 

Patients with severe short stature, GHD, prolactin deficiency and TSH deficiency only and isolated anterior pituitary findings on MRI should raise suspicion of POU1F1 mutation, especially if there is a known family history.

 

PROP1

 

The prophet of POU1F1 (PROP1) is a paired like homeodomain transcription factor on chromosome 5q35 with autosomal recessive inheritance (104). It has been shown to cause 11% of all CPHD patients, with up to 50% of familial forms and approximately 7% of sporadic forms (50). This is highly variable across different populations and ethnicities with no cases found in studies in Japan, Germany, the United Kingdom, Spain or Australia and numbers in Lithuania being as high as 65% of CPHD cases and >90% of familial cases (50).

 

Prop1 is initially expressed in Rathke’s pouch at E10, with levels rising until E12 and decreasing to E15.5 when it ceases. It stimulates stem cell transformation to mesenchymal cells and supports cell migration and differentiation, without which cells remain static. It is essential for POU1F1 initiation and failure of this results in further issues with cell differentiation. It is involved in the stimulation and differentiation of all cell lines including gonadotrophins and to a lesser extent, corticotrophs (104).

 

Most cases have CPHD, and is typically progressive, with the number of diagnosed deficiencies and severity of deficiency increasing over time. Onset of GHD, TSHD and HH can occur antenatally with diagnosis at birth, but often present later, and in rare cases, some cell lines are normal. ACTH deficiency has been described from the age of 6 and typically develops across late childhood and adolescence. Imaging demonstrates a normal or hypoplastic anterior pituitary (occasionally hyperplastic) with normal posterior pituitary structures, and aside from features secondary to hypogonadism (micropenis and undescended testis) no extracranial phenotype has been described (105).

 

The presentation is typically milder than POU1F1 mutation and the presence of features of hypogonadism and adrenal insufficiency in a non-syndromic child with CPHD should raise suspicion for PROP1 over POU1F1.

 

ISOLATED HORMONE DEFICIENCIES

 

The presence of any pituitary hormone deficiency raises the question of other hormone deficiencies. While isolated forms of pituitary deficiency occur, there is the potential for other deficiencies to develop over time as is seen in POU1F1 or LHX, and routine monitoring is required (53,100). It is also true that genes classically causing isolated hormone deficiencies, like GH1, can cause CPHD, and thus screening may be required (106,107).

 

Isolated Growth Hormone Deficiency

Figure 8. Cross-sectional representation of pituitary cell groups in isolated growth hormone deficiency. Adapted from Larkin S. et al, Endotext (1).

Isolated growth hormone deficiency (IGHD) is reported to have a prevalence between 1 in 4,000-10,000. Whilst most cases are sporadic, suggesting an in-utero insult to development of a specific cell-line, 3-30% have a first degree relative, supporting the possibility of genetic causes (108). Approximately 6.5% of patients have a genetic cause currently detectable (39) with rates as high as 38% in those with a family history (109). Growth Hormone 1 (GH1) and Growth Hormone Releasing Hormone Receptor (GHRHR) genes are the most commonly implicated, although genes implicated in CPHD, such as HESX1, SOX3, OTX2, LHX4 and GLI2 can also present as isolated GHD, (appearance shown in figure 8). Genetic causes are classified into 4 categories, listed in table 4. Type 1A which has undetectable GH levels caused by autosomal recessive GH1 gene deletions, Type 1B caused by GH1 gene splice site mutation or other genes (such as listed above), Type 2 caused by exon skipping mutations in GH1 cause an autosomal dominant phenotype, and Type 3 is X-linked recessive, and is associated with agammaglobulinemia (108).

 

The GH1 gene located on 17q22-24consists of 5 exons and 4 introns and encodes for a 191 amino acid peptide with a molecular weight of 22kDa. Transcription of GH1 is triggered by cAMP release in response to GHRH binding to the GHRHR, and is down-regulated by somatostatin via inhibiting cAMP release, in response to receptor binding (108). This pathway is demonstrated in Figure 9. GH1 mutations cause 22.7% of familial isolated GHD, the majority being type 2 autosomal dominant mutations (109).

Figure 9. Cellular function of somatotroph cell. cAMP- Cyclic Adenosine Monophosphate, GH – Growth Hormone, GHRH – Growth Hormone Releasing Hormone, GHRHR – Growth Hormone Releasing Hormone Receptor.

TYPE 1A

 

In 1981 Phillips et al. described GH1 mutation in Swiss children with a homozygous deletion causing Type 1A GHD (110). Type 1A GHD is caused by a GH1 gene deletion or less commonly, by a frameshift or nonsense mutation and makes up <15% of GH1 mutation related disease (109). The resulting phenotype is of severe GHD, with undetectable levels and can present with hypoglycemia neonatally as well as growth failure in the first 6 months. Complicating therapy is the frequent occurrence of anti-GH antibodies requiring therapy with recombinant insulin-like growth factor 1 (IGF1) (108).

 

TYPE 1B

 

Type 1B GHD caused by GH1 mutations are typically the result of gene splice site mutations and are again autosomal recessive. They have a variable phenotype ranging from an IGHD Type 1A phenotype to mid-childhood growth failure (108).

 

GHRHR anomalies also cause a Type 1B phenotype. GHRHR is found on chromosome 7p14.3 and was reported in 1996 by Wajnrajch et al. (111). Patients have normal GHRH levels but undetectable GH and low IGF1 levels. The phenotype is of proportional short stature, with anterior pituitary hypoplasia (108). GHRHR mutations are responsible for 16-19% of familial IGHD but are rarely the cause of sporadic IGHD (109). No GHRH mutations causing short stature have been reported to date (108).

 

TYPE 2

 

Type 2 GHD originates from mutations that result in loss of exon 3 from the final protein. This causes a dominant negative effect on secretion of normal GH from the other allele, through retention in the endoplasmic reticulum, impairing Golgi apparatus activity (108). Type 2 GHD has varying degrees of severity as well as of pituitary hypoplasia and represents approximately 70% of GH1 mutation related disease (109).

 

TYPE 3

 

IGHD has been reported in 10 cases of X-linked agammaglobulinemia, known as IGHD type 3; however, an individual gene causing both associations has not been well characterized (112).

 

Sporadic IGHD does not typically have a genetic basis; however, familial forms have a high likelihood of detecting a mutation (38%) (107). It is important in patients with IGHD to always consider the possibility of a developing CPHD, with this occurring in 5.5% without structural anomalies, and 20.7% with structural anomalies(106,107(113)). This should be considered even in patients with GH1 mutation’s, in particular Type 2 mutations have been found to develop other deficiencies, in particular TSH and ACTH deficiency (106,107).

 

Table 4. Summary of Types of Isolated Growth Hormone Deficiency.

IGHD type

Genetic abnormalities

Inheritance

Pituitary imaging

Percentage of familial cases

Comments

1A

GH1 gene deletions and nonsense mutations

AR

Normal/ hypoplastic

2-4%

GH levels undetectable, anti-GH antibodies, transient response to GH therapy,

1B

GH1 gene splice site mutations

AR

Normal/ hypoplastic

2-4%

 
 

GHRHR gene mutations

AR

Normal/ hypoplastic

16-19%

 

2

GH1 gene splice site and missense mutations and intronic deletions

AD

Normal/ hypoplastic

15-22%

Variable height, CPHD may occur

3

Unknown

XR

EPP

Unknown

Agammaglobulinemia

AD – Autosomal Dominant, AR – Autosomal Recessive, CPHD – Combined Pituitary Hormone Deficiency, EPP – Ectopic Posterior Pituitary, GHRHR – Growth Hormone Releasing Hormone Receptor, IGHD – Isolated Growth Hormone Deficiency, XR – X-li linked Recessive. Adapted from Alatzoglou K. et al, JCEM;94:3191-3199 and Casteras A. et al, Endocrinology, Diabetes and Metabolic Case Reports (109,114).

 

Central Hypothyroidism

 

Neonatal screening studies using T4 testing determined neonatal central hypothyroidism to occur in 1 in 16000-30000 infants (115,116). Up to 40% of these cases have isolated central hypothyroidism, with neonatal rates between 1 in 40,000 to 75,000, while some cases of isolated central hypothyroidism develop after the neonatal period (117). Thyroid hormone receptors (THR) are present on both the hypothalamus and pituitary and downregulate hormone secretion. Thyrotropin releasing hormone (TRH) is released from the hypothalamus and binds the TRH receptor (TRHR) upregulating TSH production (pathway demonstrated in Figure 10). 5 different genes have been implicated but there is limited data on the frequency of genetic mutations in patient with isolated central hypothyroidism (118).

Figure 10. Cellular function of thyrotroph cell. cAMP- Cyclic Adenosine Monophosphate, TRH – Thyrotropin Releasing Hormone, TRHR – Thyrotropin Releasing Hormone Receptor, TSH – Thyroid Stimulating Hormone.

 

TSHB

 

Thyroid stimulating hormone beta subunit (TSHB) gene is found on chromosome 1p13.1 and is associated with autosomal recessive inheritance (118). Hayashizaki et al. described the mutation in 1989 in a patient with severe neonatal onset hypothyroidism associated with prolonged jaundice, failure to thrive and developmental delay (119). Subsequent cases have also had severe deficiencies presenting neonatally (118).

 

TRHR

 

The thyrotropin releasing hormone receptor (TRHR) gene is found on chromosome 8q23.1 and again has autosomal recessive inheritance. The phenotype is milder than for TSHB with detection ranging from neonatal period to late childhood, and other patients being asymptomatic in adulthood. This is likely due to the presence of thyroid hormone receptors on the thyrotroph cells, upregulating TSH release independently of TRH (118).

 

IGSF1

 

Immunoglobulin superfamily 1 (IGSF1) gene is found on chromosome Xq26.1 and is associated with X-linked recessive central hypothyroidism. Hypothyroidism is seen in all cases, but the degree of deficiency can be variable, with many presenting in the neonatal period with jaundice and failure to thrive, others are detected asymptomatically on family screening (118).

 

IGSF1 is expressed in Rathke’s pouch and the adult pituitary gland and mutation results in low TSHB and TRHR mRNA, thus it is proposed IGSF1 has a role in regulating expression of these genes. 60% of patients have prolactin deficiency, supporting the involvement of decreased TRHR activity. GHD has been reported in a few cases, and 1 patient has been reported to have a hypoplastic anterior pituitary and ectopic posterior pituitary (118).

 

Other known associations include macroorchidism in 80% with low adulthood testosterone levels but preserved fertility. Follow up shows that despite thyroxine replacement, 25% of children and 75% of adults are overweight and 65% of cases have ADHD (attention deficit hyperactivity disorder) (118).

 

OTHER X-LINKED CAUSES

 

Transducing b-like protein X-linked (TBL1X) gene is found at chromosome Xp22.2 and mutations are thought to prevent upregulation of TSHB and TRHR transcription, causing central hypothyroidism. It has a mild hypothyroid phenotype and is associated with hearing loss, and ADHD (118).

 

Insulin receptor substrate 4 (IRS4) is found on chromosome Xq22.3 is often described as a cause of TSHD; however, mutations to date have resulted in low T4 but normal T3 and TSH, which does not suggest true central hypothyroidism (118).

 

Central hypothyroidism is rare and while genetic testing may be appropriate, the prevalence of individual causative genes is unknown. If genetics are not performed, siblings of patients should have formal thyroid function tests including a T4 arranged in the newborn period.

 

Table 5. Genetic Causes of Isolated Central Hypothyroidism

Gene

Inheritance

Degree of hypothyroidism

Other pituitary deficiency

Other features

TSHB

1p13.1

AR

Severe

Nil

Jaundice, FTT, developmental delay

TRHR

8q23.1

AR

Mild

Prolactin

 

IGSF1

Xq26.1

XR

Variable (mild to severe)

Prolactin, (rarely GH)

Macroorchidism, low testosterone, obesity, ADHD

TBL1X

Xp22.2

XR

Mild

Nil

Hearing loss, ADHD

 

AD – Autosomal Dominant, ADHD – Attention Deficit Hyperactivity Disorder, AR – Autosomal Recessive, XR – X-linked Recessive. Adapted from Tajima T. et al, Clin Pediatr Endocrinol;28(3):69-79 (118).

 

Isolated ACTH Deficiency

 

Isolated ACTH deficiency (IAD) is an extremely rare but serious condition with mortality rates of up to 25% in severe neonatal-onset forms (120).

 

Corticotrophin releasing hormone (CRH) is produced by the hypothalamus in response to low cortisol levels and binds to corticotroph cells. CRH stimulates cAMP release triggering protein kinase A (PKA) which binds CRH response element-binding protein (CREB) which in turn stimulates TBX19 which stimulates production of pro-opiomelanocortin (POMC). This is converted by prohormone convertase 1/3 (PCSK1 gene) to ACTH, which stimulates glucocorticoid production in the adrenal gland (Figure 11) (121). Abnormalities in TBX19, POMC and PCSK1 genes have all been shown to cause IAD, and NFKB2 mutation causes IAD and immunodeficiency. Genetic causes are summarized in Table 6.

Figure 11. Cellular function of the corticotroph cell. ACTH – Adrenocorticotrophic Hormone, cAMP- Cyclic Adenosine Monophosphate, CRH – Corticotropin Releasing Hormone, CRHR – Corticotropin Releasing Hormone Receptor, PC 1/3 – Prohormone Convertase 1/3, POMC – Pro-opiomelanocortin, TBX19 – T-Box Transcription Factor 19.

TBX19

 

T-box transcription factor 19 (TBX19) previously known as TPIT is located on chromosome 1q24.2 and causes severe neonatal onset IAD. It has an autosomal recessive inheritance and has been detected in 65% of cases of severe IAD, with 100% penetrance reported to date (120). As reported above, TBX19 is an important part of the pathway from CRH stimulation to ACTH release but is also critical for terminal differentiation of corticotrophs and can result in absence or severely hypoplastic cells (120).

 

All patients present with severe neonatal hypoglycemia and >50% have hypoglycemic seizures, with a mortality rate of 25%. >60% have prolonged cholestatic/ conjugated jaundice and if diagnosis is delayed, intellectual impairment and developmental delay have been reported (120). Other abnormalities have occasionally been reported including Chiari type 1 malformation, and subtle dysmorphic features (120,122).

 

Cortisol assessment should be performed in any neonate with recurrent hypoglycemia and cholestatic jaundice, and consideration of TBX19 gene assessment on confirmation of the diagnosis. Low levels of maternal estriol on antenatal testing has been shown to herald adrenal steroidogenesis issues and seen in a case of IAD caused by TBX19, but estriol is no longer routinely used in antenatal screening (123).

 

PCSK1

Proprotein convertase of Subtisilin/Kexin 1 gene (PCSK1) found on chromosome 5q15-21, codes for prohormone convertase 1/3 (PC 1/3) and has an autosomal recessive pattern of inheritance. It is found throughout the body including the pituitary, hypothalamus and endocrine pancreas. It plays a critical role in processing POMC, proinsulin and proglucagon but multiple other hormones as well (124).

 

PC 1/3 is required for conversion of POMC to ACTH in corticotroph cells. Deficiency thus results in low levels or absence of ACTH production. Despite this, there is typically a mild clinical phenotype, only 75% of patients have ACTH deficiency and not all are associated with hypocortisolemia. It is unclear whether this is due to partial ACTH binding capacity of POMC or other ACTH precursors, or whether an alternative enzyme compensates (124).

PC1/3 deficiency can also cause CPHD. HH, TSHD, and GHD are seen in approximately 50%, 70% and 35% of cases respectively and have been demonstrated or proposed to be secondary to failure of hypothalamic conversion of pro-releasing hormones (proGnRH to GnRH (suspected), proTRH to TRH, proGHRH to GHRH). Like with the corresponding adrenal insufficiency, thyroid and GH deficiency are often phenotypically mild. DI, presumed due to ineffective provasopressin conversion to vasopressin is also reported in a subset of patients (124).

 

Other associated features include obesity, occurring in nearly all patients, associated with hyperphagia and develops from approximately 2 years of age, the cause of which is multifactorial. Like in Prader-Willi syndrome, initially children present with failure to thrive; however, this is likely due to the presence of malabsorptive diarrhea. All cases to date have had severe neonatal diarrhea with demonstrated fat, amino acid and monosaccharide malabsorption, with the majority requiring hospitalization and parenteral nutrition. The underlying mechanism is not understood, and the intestinal architecture is typically normal on biopsy. The malabsorptive state typically displays partial improvement after the first year of life (124).

 

PC1/3 is also critical for cleavage of proinsulin to insulin, resulting in undetectable insulin levels. To compensate, much higher quantities of proinsulin are produced which typically prevents diabetes mellitus as it is maintains a 2-5% activity at the insulin receptor. The most common symptom is in fact postprandial hypoglycemia, resulting from proinsulins four to six times longer half-life, causing a relative hyperinsulinism after meals. Later in life some patients proceed to diabetes mellitus likely due to b-cell exhaustion (124).

 

PCSK1 mutation should be considered in patients with early onset malabsorptive diarrhea and later onset obesity. Other features may include post-prandial hypoglycemia, gonadal abnormalities and reduced linear growth. CPHD and DI should be screened for but are typically mild (124).

 

POMC

 

Pro-opiomelanocortin gene (POMC) is found on chromosome 2p23.3 and deficiency manifests in an autosomal recessive inheritance pattern (125). It causes a classic triad of adrenal insufficiency, hypopigmented skin with red hair, and obesity associated with hyperphagia, that can commence within infancy (125,126).

 

Children typically present with neonatal hypoglycemia (~70%) and adrenal insufficiency is detected at this point. Convulsions, hyperbilirubinemia and hyponatremia were present in over 30% of presenting cases. All known cases to date have been reported to have adrenal insufficiency and obesity, with 75% having red or Reddish-brown hair (125).

 

Other endocrinopathies have been reported, TSHD being the most common. Melanocyte stimulating hormones (MSH) bind the melanocortin 4 receptor which has been shown to regulate TRH release and it is hypothesized that the limited production of MSH’s drives the hypothyroidism. TSHD is reported in 35% of cases to date and is often subclinical. GHD and HH have been reported in 10-20% of patients with mechanisms not well understood. Type 1 diabetes has also been seen in 10-20% of cases (double antibody positive) it is thought this may be secondary to the increased inflammatory state in the absence of melanocortin which have been demonstrated to have an anti-inflammatory effect (125).

 

DAVID Syndrome

 

DAVID syndrome (Deficient Anterior pituitary with Variable Immune Deficiency) was first described in 2011 and results from a mutation of NFKB2 gene on chromosome 10q24.32. Classically it presents with common variable immunodeficiency (CVID) due to hypogammaglobulinemia, typically resulting in recurrent sinopulmonary infections. IAD typically develops years or decades later; however, initial presentation with IAD has been reported in 1 case (127). GHD has been reported in 2 cases and TSHD in a single case, and imaging may demonstrate a normal or hypoplastic anterior pituitary (127,128).

 

Neonatal onset isolated adrenal deficiency should be screened for TBX19 mutations, and if absent, especially in a child with red hair, POMC gene testing should be considered, similarly the possibility of PCSK1 mutation in children with malabsorptive diarrhea and adrenal insufficiency.

 

Table 6. Summary of Genetic Causes of Isolated ACTH Deficiency (IAD).

Gene

Inheritance

Onset of IAD

Presentation

Other pituitary deficiencies

Other phenotype

TBX19
Chr 1q24.2

AR, 100% penetrance

65% of severe neonatal onset IAD

Neonatal hypoglycemia, cholestatic jaundice

 Nil

Intellectual impairment if delayed diagnosis, Chiari type 1 malformation

PCSK1
Chr 5q15-21

AR

Variable

Infantile malabsorptive diarrhea,

FSH, LH, GH, TSH, Rarely ADH

Obesity in early childhood

POMC

Chr 2p23.3

AR

Neonatal

Neonatal hypoglycemia, jaundice

TSH (35%), GH, LH/FSH (10%)

Obesity, reddish brown hair. Type 1 diabetes

NFKB2

Chr 10q24.32

 

AD

Variable

Recurrent sinopulmonary infections

GH, TSH (rare)

Combined variable immunodeficiency

AD -- Autosomal Dominant, AR – Autosomal Recessive.

 

Central Hypogonadism

 

Congenital hypogonadotrophic hypogonadism (HH), also known as Kallmann syndrome when seen in association with anosmia, occurs in 1 in 10,000 – 50,000 people, with a 4 to 1 male predominance (12,129).

 

Gonadotrophin releasing hormone (GnRH) is secreted in a pulsatile manner from the hypothalamus and has a complex pattern of regulation by estradiol and other factors. It binds GnRH receptors on gonadotroph cells leading to secretion of LH and FSH. In fetal development, the cells responsible for GnRH release migrate to the hypothalamus from the olfactory placode, and thus mutations triggering HH are often associated with olfactory dysgenesis and anosmia (12).

 

There are over 25 gene defects implicated in HH, and a mutation is found in up to 50% of cases. No gene is implicated in more than 10% of familial cases. Oligogenicity is also described in multiple genes which further complicates diagnosis. Broadly speaking, HH is a result of one of four mechanisms, defects in GnRH fate specification, GnRH neuron migration, abnormal neuroendocrine secretion/homeostasis or gonadotroph cell defects (130).

 

55% of cases have anosmia/hyposmia (129). The majority of anosmic cases have an autosomal dominant inheritance, except for FEZF1, PROK2 and PROKR2, while norosmic cases may be autosomal dominant or recessive. However, incomplete penetrance, oligogenicity and variability of anosmia and norosmia of some genes makes delineating likely causative genes difficult. X-linked recessive inheritance is seen in KAL1 mutations only to date (12,130).

 

While individual genes have specific associations, larger studies demonstrate between 5 and 15% of cases have hearing loss, palate anomalies or unilateral renal agenesis. Up to 30% have synkinesia or eye movement disorders and one study found 5-15% of patients had dental agenesis, scoliosis or finger anomalies (129).

 

In the absence of anosmia or congenital anomalies, HH can be difficult to separate from constitutional delay (normal late onset puberty). Adolescents require pubertal induction to support bone, metabolic and sexual health and to limit the effect on mental health, and input from fertility specialists may be required when starting a family (129).

 

Multiple genes associated with HH have been implicated in combined hypopituitarism including PROKR2, FGF8, FGFR1 and CHD7 and summarized in Table 7.

 

CHARGE Syndrome (CHD7)

 

CHARGE syndrome results from mutation of the CHD7 gene on chromosome 8q12.2 with autosomal dominant inheritance, although typically resulting from de novo mutation and occurs in 1 in 15,000 to 17,000 live births (131). The acronym CHARGE is of Coloboma of the eye, Heart defects, Atresia of the choanae, Retardation of growth and development, Genital hypoplasia and Ear and hearing abnormalities, and was coined by Pagon et al. in 1981 (132).

 

HH, typically presenting with cryptorchidism and micropenis in males is present in 60-88% of patients, most of whom will not enter puberty spontaneously (131,133). Growth failure is present in 60-72% of cases, with approximately 10% being due to GHD (131,134-136). Other pituitary abnormalities are rare with one study reporting 2 out of 25 patients with TSHD, and while other studies report hypothyroidism, these are typically primary, or the etiology is not reported. Imaging typically demonstrates olfactory bulb anomalies, but the pituitary gland is typically normal (134,136). Thus, while hypogonadism is a well-known consequence of CHARGE syndrome, GHD and TSHD must also be considered.

 

PROKR2

 

Prokineticin-2 Receptor (PROKR2) gene is found on chromosome 20p13 and typically has dominant inheritance with incomplete penetrance, but recessive mutations are also described (70). PROKR2 prevalence within HH varies greatly, ranging from 5.1-23.3% of mutations in different populations(137). PROKR2 is highly expressed in the olfactory placode and GnRH neurons in the mouse model but is not seen in the pituitary or infundibulum. It typically causes HH and individuals often harbor mutations in other HH genes, demonstrating it sometimes has an oligogenic effect (70). CPHD has been described in 20 patients, 13 of whom had SOD and 4 with pituitary stalk interruption syndrome (PSIS) (5-8). The pathogenicity of the mutations in these cases is uncertain (6,70).

 

FGF8

 

The fibroblast growth factor 8 (FGF8) gene is found on chromosome 10q24 and mutations can have either autosomal dominant or recessive inheritance, making up approximately 1% of HH cases (70,137). FGF8 is first expressed at E9.25 in the diencephalon, prospective hypothalamus and infundibulum, and activates LHX3 (70,96,138,139). FGF8 plays a role in coordinating anterior pituitary development, with excess in mice resulting in a dysplastic, hyperplastic pituitary and absence results in pituitary hypoplasia. Most mutations are associated with Kallmann syndrome; however, multiple cases of CPHD have been identified as well as a case of Holoprosencephaly, and SOD with GH deficiency. VACTERL (Vertebral defects, Anal atresia, Cardiac defects, Tracheo-esophageal fistula, Renal anomalies, and Limb abnormalities) or VACTERL like phenotype has also been described (70).

 

FGFR1

 

The Fibroblast Growth Factor Receptor 1 (FGFR1) gene is found on chromosome 8p11.2-p12 with autosomal dominant inheritance making up 5-11.7% of mutations associated with HH (138,140). It is expressed in the olfactory placode where is ensures fate specification, proliferation and migration of GnRH neurons (140). It typically causes HH, with or without anosmia but also been reported in CPHD. 9 cases have been described, 3 associated with SOD, 3 with PSIS and 3 without (5,7,141,142). While some variants have demonstrable pathogenicity for this phenotype, it is unclear in others. Split hand/foot malformation is frequently part of the FGFR1 phenotype and the mutation is also seen in Pfeiffer syndrome, Hartsfield syndrome and Jackson-Weiss syndrome (142).

 

Table 7. Causes of Hypogonadotrophic Hypogonadism Associated with Combined Pituitary Hormone Deficiency.

Gene

Inheritance/ prevalence

SOD/

PSIS/HPE

Other pituitary deficiencies

Other Features

PROKR2

Chr 20p12.3

AD/AR

5-23% of HH

SOD/ PSIS

CPHD

 

FGF8

Chr 10q24

AD/AR

1% of HH

SOD/HPE

GH/CPHD

VACTERL

FGFR1

Chr 8p11.2-p12

AD

5-11% of HH

SOD/PSIS

GH/CPHD

Split hand/foot malformation

CHARGE

CHD7

Chr 8q12.2

AD/de novo

6% of HH

Nil

GH/TSH

Coloboma, CHD, choanal atresia, short stature, ID, genital hypoplasia, ear abnormalities

 AD – Autosomal Dominant, AR – Autosomal Recessive, CHD – Congenital Heart Disease HPE – Holoprosencephaly, ID – Intellectual Disability, PSIS – Pituitary Stalk Interruption Syndrome, SOD – Septo-optic Dysplasia, VACTERL- Vertebral defects, Anal atresia, Cardiac defects, Tracheo-esophageal fistula, Renal anomalies, and Limb abnormalities.

 

Central Diabetes Insipidus

 

Isolated diabetes insipidus (DI) is rare, with a reported prevalence of 1 in 25000, including both nephrogenic and central forms (143). Arginine vasopressin (AVP) or antidiuretic hormone (ADH) is produced from pre-pro-AVP in the supraoptic and paraventricular hypothalamic nuclei, and their axonal projections extend into the posterior pituitary. AVP mediates its affect through the AVPR2 receptor which increases urine concentration in the collecting duct. Apart from cases related to holoprosencephaly, genetic causes are rare (144).

 

AVP

 

The AVP gene is located on chromosome 20p13 and has an autosomal dominant inheritance except for a few recessive mutations. The AVP mutant proteins are thought to accumulate in the endoplasmic reticulum resulting in cell death. As a result, it usually manifests between 1 to 6 years of age with gradual progression of deficiency but there is wide variation even within affected families. Along with polyuria and polydipsia classically seen in DI, failure to thrive is commonly seen and resolves with AVP replacement (144).

 

Wolfram Syndrome

 

The WFS1 gene, located on chromosome 4p16.1 encodes for wolframin which is an endoplasmic reticulum channel and has an autosomal recessive inheritance (144). Prevalence is estimated between 1 in 500,000 – 770,000 children with rates higher in areas of increased consanguinity (1 in 68,000 in Lebanon) (145). Wolfram syndrome causes diabetes mellitus, usually diagnosed at 6 years of age, optic nerve atrophy at around age 11 and sensorineural hearing loss typically progressing over time. DI is seen in approximately 70% of cases and is typically diagnosed in the 2nd or third decade of life (143). A case DI without other manifestations of Wolfram syndrome has been reported (146). The mechanism of DI in Wolfram syndrome is not well understood (144).

 

SYNDROMES ASSOCIATED WITH HYPOPITUITARISM

 

Hypopituitarism has been reported in a wide variety of syndromes as an associated feature. These syndromes are reported separately as hypopituitarism is a less common association.

 

Prader-Willi Syndrome

 

Prader Willi Syndrome (PWS) is a complex condition with major neurological and endocrine challenges. It results from a lack of the paternally imprinted genes on chromosome 15q11.2-q13 which can result from paternal gene deletion, maternal uniparental disomy, or imprinting defects (147). Children typically have hypotonia and failure to thrive in infancy, but later progress to hyperphagia and obesity. Obesity and pituitary hormone deficiencies result from hypothalamic dysfunction. Growth failure and GHD are commonly seen but CPHD can also occur, and the anterior pituitary can be hypoplastic in approximately 70% of patients (148,149).

 

Growth failure in Prader-Willi Syndrome occurs due to failure within the GH-IGF-1 axis, with GHD reported in 40-100% of patients and IGF-1 deficiency in nearly 100% of cases. Growth hormone replacement early in life (<1 year of age) is recommended regardless of GHD and is shown to improve lean muscle mass, motor development and cardiovascular health (150-153).

 

Primary gonadal failure is present in most cases, with underdeveloped gonads typical in both genders. HH may compound this in some cases (154). TSHD has been reported in up to 30% cases, but the true prevalence is unclear. High rates were reported in the population younger than 2 years of age but in older children, rates did not exceed the general population, suggesting CNS maturation may result in this early childhood phenotype being transient. It is unclear if there is an association between PWS and ACTH deficiency with reported rates ranging from 0-60%, with rates likely being 5-14%. Routine screening of 8 AM cortisol is recommended (147).

 

The mechanism for hypopituitarism in PWS is not well understood. The MAGEL2 gene, located within the PWS locus causes Schaaf-Yang Syndrome and presents similarly to PWS but with arthrogryposis. MAGEL2 is highly expressed in the developing hypothalamus and pituitary between 7-8 weeks gestation and mutations can result in IGHD or CPHD (155).

Genetic testing is indicated in children with a history of neonatal hypotonia and failure to thrive that progress to obesity, with or without short stature.

 

Axenfield-Rieger Syndrome (PITX2)

 

Axenfield-Rieger syndrome involves dysgenesis of the anterior segment of the eye, typically resulting in glaucoma. The cause of many cases is unknown; however, 40% are due to either the PITX2 or FOXC2 genes. PITX2 is a pituitary homeobox gene located on chromosome 4q25 with autosomal dominant inheritance and variable penetrance (156). PITX2 is expressed early in the oral ectoderm (E8.5), and later in the anterior pituitary where it induces POU1F1 amongst other factors (156,157). Some cases of PITX2 mutation present with GHD, with dental and cardiac issues also associated (156).

 

Johanson-Blizzard Syndrome (UBR1)

 

Mutation of the UBR1 gene on 15q15-21 results in Johanson-Blizzard syndrome (JBS) and is inherited autosomal recessively, occurring in 1 in 250,000 live births, with 70 patients reported in the literature (158). It is characterized by exocrine pancreatic insufficiency, nasal alae hypoplasia/aplasia and cutis aplasia of the scalp. Along with developmental delay, failure to thrive and hearing impairment, hypothyroidism, genitourinary abnormalities and hypopituitarism are sometimes reported (159).

 

In 2007, Hoffman et al. reported a case of a 4-year-old deceased boy with ACTH, GH and TSH deficiency, with undescended testicles and hypoplastic pituitary gland with an absent stalk (160). 3 cases of IGHD and isolated TSHD have also been reported, with microgenitalia reported in 4 patients, who have not been evaluated for HH. Other known endocrine associations include diabetes mellitus and primary hypothyroidism (158,159).

 

Oliver-McFarlane Syndrome (PNPLA6)

 

Oliver McFarlane syndrome is caused by a recessive mutation of chromosome 19p13 encoding for the PNPLA6 gene. This encodes for neuropathy target esterase (NTE) and is found in the human eye, brain and pituitary, which also results in other syndromes including Gordon-Holmes, Lawrence-Moon and Boucher–Neuhäuser syndrome, which are associated with HH and occasionally hypopituitarism (161). A 2021 systematic review found of the 31 cases in the literature, 90% had hypopituitarism, with 67% having GHD or HH and 50% having TSHD (162). Other features include chorioretinal atrophy, trichomegaly, alopecia, and spinocerebellar involvement (161,162).

 

Wiedemann-Steiner Syndrome (KMT2A)

 

Wiedemann-Steiner syndrome results from heterozygous mutation of the KMT2A gene on chromosome 11q23.3. It classically presents with intellectual disability, facial dysmorphism, hypertrichosis of the elbow and short stature. A study of 104 individuals found short stature in 57.8% (163) and of the 12 patients tested within a cohort of 33, 6 were found to have GHD. The mechanism for hypopituitarism is not well understood (164).

 

Kabuki Syndrome (KMT2D)

 

Mutations of KMT2D gene on chromosome 12q13.12 are responsible for 55-80% of cases of Kabuki syndrome and result in autosomal dominant inheritance (165). Kabuki syndrome is characterized by distinctive facial features, intellectual disability, short stature and cardiac disease. 10 cases of hypopituitarism have been reported in the literature, 6 with GHD, 2 with DI and precocious puberty, 1 with ACTH deficiency and 1 with GHD and DI. Imaging showed 2 had PSIS and 1 had an absent posterior pituitary bright spot (166).

 

Williams Syndrome

 

Deletions of the 7q11.23 region result in Williams or Williams-Beuren syndrome, which has autosomal dominant inheritance. Williams syndrome occurs in between 1 in 7,500 and 20,000 live births and is characterized by congenital heart disease (supravalvular aortic stenosis), intellectual impairment, short stature and facial dysmorphism (167). 40% of patients have short stature, with hypercalcemia, precocious puberty and primary hypothyroidism commonly reported (168). Growth hormone deficiency is less frequently reported (168-170), and a case of combined ACTH and TSH deficiency has also been described (167). MRI imaging is typically normal, with one patient having an absent posterior-pituitary bright spot but normal ADH production (170), and another with ONH and normal pituitary function has been described (171).

 

PHACES

PHACES syndrome is a rare constellation of signs including Posterior fossa brain malformations, cervicofacial infantile Hemangiomas, Arterial anomalies, Cardiac defects, Eye anomalies and midline/ ventral defects. It is typically diagnosed following the detection of large, segmental facial hemangiomas and no gene defect has been identified. Over 300 cases have been reported to date (172). Goddard et al. reported a child with partially empty sella and GHD and TSHD in 2006 (173). Since then, 11 cases with hypopituitarism have been reported, 7 with abnormal pituitary MRI, 2 unreported, and 2 with normal pituitary appearance. The majority had GH (9/11) or TSH deficiency (7/11) with adrenal insufficiency and HH occurring in 2 out of 11 each. There is also an association with thyroid development issues and primary hypothyroidism (172). Thus, patients with PHACES syndrome should be monitored for growth and development with consideration of endocrine assessment if there are concerns. Abnormal pituitary appearance on MRI requires routine endocrine follow up (174).

 

Table 8. Syndromes Associated with Hypopituitarism.

Gene

Inheritance/ prevalence

Pituitary appearance

Pituitary deficiencies

Other Features

Prader Willi

15q11.2-q13

Imprinted

Normal

GH,

TSH, FSH/LH, ACTH

Infantile FTT, obesity, hyperphagia, ID, typically primary hypogonadism, dysmorphic features

Axenfield-Rieger PITX2

Chr 4q25

AD

Normal

GH

Eye changes, glaucoma, dental issues, CHD

Johansson Blizzard

UBR1

15q15-21

AR

APH, absent stalk

GH, TSH,

ACTH
?FSH/LH

Exocrine pancreas insufficiency, nasal anomalies, cutis aplasia, ID, genitourinary anomalies

Oliver-McFarlane

PNPLA6

Chr 19p13

AR

 

Normal

GH, FSH/LH, TSH

Chorioretinal atrophy, trichomegaly, alopecia, spinocerebellar involvement.

Wiedemann-Steiner

KMT2A

Chr 11q23.3

AR

Normal

GH

ID, facial dysmorphism, elbow hypertrichosis, short stature

Kabuki

KMT2D

Chr 12q13.12

AD

Normal, PSIS / absent PP

GH,

ADH, ACTH

Facial dysmorphism, ID, short stature, CHD

Williams

Chr7q11.23

AD

Normal

GH, TSH, ACTH

Facial dysmorphism, ID, short stature, CHD, hypercalcemia, precocious puberty, primary hypothyroidism

PHACES

(gene unknown)

Unknown

APH

GH, TSH, ACTH, FSH/LH

Posterior fossa brain malformations, hemangiomas, arterial anomalies, CHD, eye anomalies and midline/ ventral defects.

ACTH – Adrenocorticotrophic Hormone, AD – Autosomal Dominant, APH – Anterior Pituitary Hypoplasia, CHD – Congenital Heart Disease, FTT – Failure To Thrive, ID – Intellectual Disability, PP – Posterior Pituitary, PSIS – Pituitary Stalk Interruption Syndrome.

 

INDICATIONS FOR GENETIC TESTING

 

Known genetic causes make up only a small proportion of the current hypopituitarism population, thus routine broad screening will not be appropriate at most centers and currently remains in the realms of research. Testing should be targeted to patients with consistent features, such as anophthalmia and SOX2 or a short stiff neck in LHX4. Individual genes can have a wide range of presentations and thus presence of a family history of hypopituitarism, whether similar in presentation or varied, should raise a high index of suspicion and screening should be strongly considered. Autosomal recessive causes are frequently seen, and dominant causes often have incomplete penetrance thus even relatively distant family associations should cause consideration for testing. Finally, presence of hypopituitarism in the context of a consanguineous parents should again raise suspicion of a likely genetic cause. Genetic testing typically is through next generation sequencing (NGS), although some mutations may be detectable on chromosomal microarray. Deletions on chromosomal microarray may include associated genes causing hypopituitarism. All the genes described above are summarized in table 9, in order of gene location, to assist in determining which mutations may be associated in such cases.

 

Septo-Optic Dysplasia

 

Current evidence suggests genetic causes of SOD make up <10% of cases thus routine screening will not be appropriate in most centers (19). In the absence of family history, the main indication for testing is in patients with microphthalmia or anophthalmia, where testing for SOX2 and OTX2 would be appropriate, and if negative, testing could be extended to PAX6, RAX and BMP4 (38). In patients with facial midline defects or polydactyly, GLI2 testing should also be considered (66).

 

Syndromic Causes

 

Syndromic causes should be tested for based on clinical suspicion, with family history and consanguinity again increasing the likelihood. Respiratory distress syndrome and a short stiff neck or Chiari I malformation should raise suspicion of LHX3 and LHX4 mutations respectively (53,56). Polydactyly and either midline facial changes or hypothalamic hamartoma suggest GLI2 and GLI3 mutations respectively (66,68). In cases of holoprosencephaly with anterior pituitary dysfunction, outside of GLI2, there are only case reports of other known genes causing hypopituitarism (66).

 

Non-Syndromic

 

POU1F1 testing may be appropriate in patients with severe short stature, GHD, TSHD and prolactin deficiency alone, especially in Western-Indian populations where rates of up to 25% of the CPHD cohort have been reported (100). PROP1 typically has progressive hypopituitarism with late onset ACTH deficiency and routine screening in European countries, particularly Lithuania, may be appropriate (50).

 

Isolated Hormone Deficiencies

 

Unless familial, isolated GH deficiency rarely has a genetic basis thus genetic testing is not currently indicated with the exception of those with absent GH levels on stimulation testing as seen in type 1a GH1 mutations (109). The frequency of genetic causes within the TSH deficient cohort is unknown. Macroorchidism points to IGSF1 mutation but otherwise family history is the main indicator for genetic testing (118). Cases with isolated ACTH deficiency in the neonatal period should be tested for TBX19 mutations, and if absent, especially in a child with red hair, POMC gene testing should be performed (120,124). Children with malabsorptive diarrhea and adrenal insufficiency should be screened for PCSK1 mutation, and NFKB2 if there is known immunodeficiency (125,127). Hypogonadotrophic hypogonadism testing is well described and typically involves panel testing, but targeted testing based on inheritance pattern may be appropriate (12,130). Isolated diabetes insipidus is rare and testing should be based on a family history or presence of other features consistent with Wolfram syndrome, such as childhood onset optic atrophy or diabetes mellitus (144).

 

Syndromes Associated with Hypopituitarism

 

Testing for Prader-Willi, Williams, PITX2, UBR1, PNPLA6, KMT2A and KMT2D gene mutations should be considered based on their clinical features as listed in table 8, and in patients known to have these mutations, pituitary screening may be necessary, especially in the context of growth failure.

 

Table 9. Summary of Genes Associated with Hypopituitarism

Gene

Inheritance

Pituitary deficiencies

Phenotype

1p13.1 - TSHB

AR

TSH

Jaundice, FTT, developmental delay

1q24.2 - TBX19

AR,

 ACTH

Intellectual impairment if delayed diagnosis, Chiari type 1 malformation 

1q25.2 - LHX4

AD

CPHD

CC hypoplasia, Chiari 1 malformation, rarely CHD/RDS

2p11.2 - TCF7L1

AD

GH, ACTH

SOD, CC/SP changes

2p21 - SIX3

AD

CPHD

HPE, PSIS

2p23.3 - POMC

AR

ACTH, TSH (35%), GH, LH/FSH (10%)

Obesity, reddish brown hair. Type 1 diabetes

2q14.2 - GLI2

AD

GH, CPHD

SOD, PSIS, SP/CC changes, polydactyly, midface hypoplasia, cleft palate/lip, HPE

3p11.2 - POU1F1

AR/AD

GH, TSH, prolactin (typically CPHD)

 

3p12.3 - ROBO1

AD/AR

GH, TSH, CPHD

PSIS, hypermetropia, ptosis

3p14.3 - HESX1

AD/AR

GH, CPHD, (ACTH, ADH uncommon)

SOD, CC/SP changes, hypoplastic olfactory, underdeveloped forebrain

3q26.33 - SOX2

AD

GH, LH/FSH

SOD, AO/MO, hypothalamic hamartoma, CC changes, SNHL, ID,
esophageal anomalies, genital anomalies

4p16.1 - WFS1 Wolfram

AR

ADH

Diabetes mellitus, optic nerve atrophy, SNHL

4q25 - PITX2 Axenfield-Rieger

AD

GH

Eye changes, glaucoma, dental issues, CHD

5q15-21 - PCSK1

AR

ACTH, FSH, LH, GH, TSH, Rarely ADH

Obesity in early childhood

5q35 - PROP1

AR

CPHD

 

6q22.31 - TBC1D32

AR

GH, CPHD

SOD, Oro-facial-digital syndrome, CC agenesis

7p14.1 - GLI3 Pallister-Hall

AD

CPHD

Hypothalamic hamartoma, Postaxial polydactyly, imperforate anus. CPP

7p14.3 - GHRHR

AR

GH

 

7q11.23 - Williams

AD

GH, (TSH, ACTH uncommon)

Facial dysmorphism, ID, short stature, CHD, hypercalcemia, precocious puberty, primary hypothyroidism

7q36 - SHH

AD

CPHD

HPE, PSIS, renal-urinary anomalies

8p11.2-p12 - FGFR1

AD

FSH/LH, (GH/CPHD uncommon)

SOD, PSIS, split hand/foot malformation

8q12.2 – CHD7 CHARGE

AD/de novo

FSH/LH, (GH/TSH uncommon)

Coloboma, CHD, choanal atresia, short stature, ID, genital hypoplasia, ear abnormalities

8q23.1 - TRHR

AR

TSH, Prolactin

 

9q34.3 - LHX3

AR

CPHD, ACTH in 50%

SNHL, ID, short stiff neck, scoliosis, rarely RDS

10q24.32 - NFKB2

AD

ACTH, (GH, TSH uncommon)

Combined variable immunodeficiency

10q24.32 - FGF8

AD/AR

FSH/LH, (GH/CPHD uncommon)

SOD, HPE, VACTERL

11p13 - PAX6

AD

GH, ACTH, FSH/LH

SOD, AO/MO, ASD, ADHD, obesity, diabetes mellitus

11q23.3 - KMT2A Wiedemann-Steiner

AR

GH

ID, facial dysmorphism, elbow hypertrichosis, short stature

11q24.2 - CDON

AD

CPHD

HPE, PSIS, CHD, renal dysplasia, radial defects, gallbladder agenesis

12q13.12 - KMT2D Kabuki

AD

GH, ADH, ACTH

Facial dysmorphism, ID, short stature, CHD

13q32 - ZIC2

AD

CPHD

HPE, PSIS, renal-urinary anomalies

14q22.3 - OTX2

AD

GH, CPHD,
(ACTH uncommon)

SOD, PSIS, AO/MO

14q22-23 - BMP4

AD

GH, TSH

myopia, cleft palate/lip, polydactyly

15q11.2-q13 - Prader Willi

Imprinted

GH, TSH, FSH/LH, ACTH

Infantile FTT, obesity, hyperphagia, ID, primary hypogonadism, dysmorphic features

15q15-21 - UBR1 Johansson Blizzard

AR

GH, TSH, (ACTH uncommon)

Exocrine pancreas insufficiency, nasal anomalies, cutis aplasia, ID, genitourinary anomalies

15q25.1 - ARNT2

AR

CPHD + ADH

Microcephaly, fronto-temporal hypoplasia, renal anomalies and vision impairment.

17q22-24 - GH1

AR/AD

GH, (CPHD uncommon)

Anti-GH antibodies in some forms

18p11.3 - TGIF

AD

CPHD

HPE, PSIS

18q21.32 - RAX

AR

CPHD

SOD, AO/MO, palate changes

19p13 - PNPLA6 Oliver-McFarlane

AR

GH, FSH/LH, TSH

Chorioretinal atrophy, trichomegaly, alopecia, spinocerebellar involvement.

20p11.21 - FOXA2

AD/de novo

CPHD

PSIS, hyperinsulinism, craniofacial dysmorphism, choroidal coloboma, and liver, lung and heart malformations

20p12.3 - PROKR2

AD/AR

FSH/LH, (CPHD uncommon)

SOD, PSIS

20p13 - AVP

AD/AR

ADH

FTT, polyuria

Xp22.11 - EIF2S3

XR

GH, CPHD

CC hypoplasia, ID, epilepsy, gonadal failure, microcephaly, obesity

Xp22.2 - TBL1X

XR

TSH

Hearing loss, ADHD

Xq26.1 - IGSF1

XR

TSH, Prolactin (GH uncommon)

Macroorchidism, low testosterone, obesity, ADHD

Xq27 - SOX3

XR

GH (CPHD uncommon)

PSIS, CC changes, ID

AD – Autosomal Dominant, ADHD – Attention Deficit Hyperactivity Disorder, AO – Anophthalmia, AR – Autosomal Recessive, ASD – Autism Spectrum Disorder, CC– Corpus Callosum, CHD – Congenital Heart Disease, CPHD – Combined Pituitary Hormone Deficiency, CPP – Central Precocious Puberty, FTT – Failure To Thrive, HH – Hypogonadotrophic Hypogonadism, HPE – Holoprosencephaly, ID – Intellectual Disability, MO – Microphthalmia, PSIS – Pituitary Stalk Interruption Syndrome, RDS – Respiratory Distress Syndrome, SOD – Septo-optic dysplasia, SP – Septum Pellucidum, SNHL – Sensorineural Hearing Loss, VACTERL- Vertebral defects, Anal atresia, Cardiac defects, Tracheo-esophageal fistula, Renal anomalies, and Limb abnormalities, XR – X-linked Recessive.

 

ABBREVIATION LIST

 

ACTH – Adrenocorticotrophic Hormone

AD       – Autosomal Dominant

ADH    – Antidiuretic Hormone (also known as AVP)

ADHD – Attention Deficit Hyperactivity Disorder

APH    – Anterior Pituitary Hypoplasia

αMSH – Alpha-Melanocyte Stimulating Hormone

AR       – Autosomal Recessive

AVP     – Arginine Vasopressin (also known as ADH)

CC       – Corpus Callosum

CHD    – Congenital Heart Disease

CPHD  – Combined Pituitary Hormone Deficiency

CRH    – Corticotrophin Releasing Hormone

CVID   – Common Variable Immunodeficiency

DI        – Diabetes Insipidus

EPP     – Ectopic Posterior Pituitary

FSH     – Follicle Stimulating Hormone

GH      – Growth Hormone

GHD    – Growth Hormone Deficiency

HH       – Hypogonadotrophic Hypogonadism

HPE    – Holoprosencephaly

IAD      – Isolated ACTH Deficiency

ID        – Intellectual Disability

IGF1    – Insulin-like Growth Factor 1

IGHD   – Isolated Growth Hormone Deficiency

LH       – Luteinizing Hormone

MPHD – Multiple Pituitary Hormone Deficiency

NGS    – Next Generation Sequencing

ONH    – Optic Nerve Hypoplasia

PSIS    – Pituitary Stalk Interruption Syndrome

SOD    – Septo-Optic Dysplasia

SP       – Septum Pellucidum

THR    – Thyroid Hormone Receptor

TRH    – Thyrotropin Releasing Hormone

TRHR – Thyrotropin Releasing Hormone Receptor

TSH     – Thyroid Stimulating Hormone

TSHD – Thyroid Stimulating Hormone Deficiency

XR       – X-linked Recessive

 

REFERENCES

 

  1. Larkin S, Ansorge O. Development And Microscopic Anatomy Of The Pituitary Gland. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA)2000.
  2. Sheng HZ, Westphal H. Early steps in pituitary organogenesis. Trends Genet.1999;15(6):236-240.
  3. Vimpani GV, Vimpani AF, Lidgard GP, Cameron EH, Farquhar JW. Prevalence of severe growth hormone deficiency. Br Med J.1977;2(6084):427-430.
  4. Gregory LC, Dattani MT. The Molecular Basis of Congenital Hypopituitarism and Related Disorders. J Clin Endocrinol Metab.2020;105(6):2103-2120.
  5. Correa FA, Trarbach EB, Tusset C, Latronico AC, Montenegro LR, Carvalho LR, Franca MM, Otto AP, Costalonga EF, Brito VN, Abreu AP, Nishi MY, Jorge AAL, Arnhold IJP, Sidis Y, Pitteloud N, Mendonca BB. FGFR1 and PROKR2 rare variants found in patients with combined pituitary hormone deficiencies. Endocrine Connections.2015;4:100-107.
  6. McCabe MJ, Gaston-Massuet C, Gregory LC, Alatzoglou KS, Tziaferi V, Sbai O, Rondard P, Masumoto K-h, Nagano M, Shigeyoshi Y, Pfeifer M, Hulse T, Buchanan CR, Pitteloud N, Martinez-Barbera J-P, Dattani MT. Variations in PROKR2, But Not PROK2, Are Associated With Hypopituitarism and Septo-optic Dysplasia. J Clin Endocrinol Metab.2013;98(3):547-557.
  7. Raivio T, Avbelj M, McCabe MJ, Romero CJ, Dwyer AA, Tommiska J, Sykiotis GP, Gregory LC, Diaczok D, Tziaferi V, Elting MW, Padidela R, Plummer L, Martin C, Feng B, Zhang C, Zhou QY, Chen H, Mohammadi M, Quinton R, Sidis Y, Radovick S, Dattani MT, Pitteloud N. Genetic overlap in Kallmann syndrome, combined pituitary hormone deficiency, and septo-optic dysplasia. J Clin Endocrinol Metab.2012;97(4):E694-699.
  8. Reynaud R, Jayakody SA, Monnier C, Saveanu A, Bouligand J, Guedj AM, Simonin G, Lecomte P, Barlier A, Rondard P, Martinez-Barbera JP, Guiochon-Mantel A, Brue T. PROKR2 variants in multiple hypopituitarism with pituitary stalk interruption. J Clin Endocrinol Metab.2012;97(6):E1068-1073.
  9. Skowronska-Krawczyk D, Rosenfield M, eds. Development of the Pituitary. 6 ed. Philadelphia: Saunders Elsevier; 2010. Jameson JL, Groot LD, eds. Endocrinology; No. 1
  10. Zhu X, Gleiberman AS, Rosenfeld MG. Molecular physiology of pituitary development: signaling and transcriptional networks. Physiol Rev.2007;87(3):933-963.
  11. Rizzoti K, Lovell-Badge R. Early development of the pituitary gland: induction and shaping of Rathke's pouch. Rev Endocr Metab Disord.2005;6(3):161-172.
  12. Millar AC, Faghfoury H, Bieniek JM. Genetics of hypogonadotropic hypogonadism. Transl Androl Urol.2021;10(3):1401-1409.
  13. Ludwig P, Lopez MJ, Czyz CN. Embryology, Eye Malformations. StatPearls [Internet].2021.
  14. Matsumoto R, Takahashi Y. Human pituitary development and application of iPSCs for pituitary disease. Cell Mol Life Sci.2021;78(5):2069-2079.
  15. Hill M. Embryology Endocrine - Pituitary Development. 2022.
  16. Mitrofanova LB, Konovalov PV, Krylova JS, Polyakova VO, Kvetnoy IM. Plurihormonal cells of normal anterior pituitary: Facts and conclusions. Oncotarget.2017;8(17):29282-29299.
  17. Patel L, McNally RJ, Harrison E, Lloyd IC, Clayton PE. Geographical distribution of optic nerve hypoplasia and septo-optic dysplasia in Northwest England. J Pediatr.2006;148(1):85-88.
  18. Garne E, Rissmann A, Addor MC, Barisic I, Bergman J, Braz P, Cavero-Carbonell C, Draper ES, Gatt M, Haeusler M, Klungsoyr K, Kurinczuk JJ, Lelong N, Luyt K, Lynch C, O'Mahony MT, Mokoroa O, Nelen V, Neville AJ, Pierini A, Randrianaivo H, Rankin J, Rouget F, Schaub B, Tucker D, Verellen-Dumoulin C, Wellesley D, Wiesel A, Zymak-Zakutnia N, Lanzoni M, Morris JK. Epidemiology of septo-optic dysplasia with focus on prevalence and maternal age - A EUROCAT study. Eur J Med Genet.2018;61(9):483-488.
  19. McCabe MJ, Alatzoglou KS, Dattani MT. Septo-optic dysplasia and other midline defects: the role of transcription factors: HESX1 and beyond. Best Pract Res Clin Endocrinol Metab.2011;25(1):115-124.
  20. Atapattu N, Ainsworth J, Willshaw H, Parulekar M, MacPherson L, Miller C, Davies P, Kirk JM. Septo-optic dysplasia: antenatal risk factors and clinical features in a regional study. Horm Res Paediatr.2012;78(2):81-87.
  21. Garcia-Filion P, Borchert M. Prenatal determinants of optic nerve hypoplasia: review of suggested correlates and future focus. Surv Ophthalmol.2013;58(6):610-619.
  22. Cerbone M, M.Guemes, Wade A, Improda N, Dattani M. Endocrine morbidity in midline brain defects: Differences between septo-optic dysplasia and related disorders. EClinicalMedicine.2020;19:1-17.
  23. Cemeroglu AP, Coulas T, Kleis L. Spectrum of clinical presentations and endocrinological findings of patients with septo-optic dysplasia: a retrospective study. J Pediatr Endocrinol Metab.2015;28(9-10):1057-1063.
  24. Oatman OJ, McClellan DR, Olson ML, Garcia-Filion P. Endocrine and pubertal disturbances in optic nerve hypoplasia, from infancy to adolescence. Int J Pediatr Endocrinol.2015;2015(1):8.
  25. Ahmad T, Garcia-Filion P, Borchert M, Kaufman F, Burkett L, Geffner M. Endocrinological and auxological abnormalities in young children with optic nerve hypoplasia: a prospective study. J Pediatr.2006;148(1):78-84.
  26. Siatkowski RM, Sanchez JC, Andrade R, Alvarez A. The clinical, neuroradiographic, and endocrinologic profile of patients with bilateral optic nerve hypoplasia. Ophthalmology.1997;104(3):493-496.
  27. McNay DE, Turton JP, Kelberman D, Woods KS, Brauner R, Papadimitriou A, Keller E, Keller A, Haufs N, Krude H, Shalet SM, Dattani MT. HESX1 mutations are an uncommon cause of septooptic dysplasia and hypopituitarism. J Clin Endocrinol Metab.2007;92(2):691-697.
  28. Kelberman D, Dattani MT. Genetics of septo-optic dysplasia. Pituitary.2007;10(4):393-407.
  29. Tajima T, Ohtake A, Hoshino M, Amemiya S, Sasaki N, Ishizu K, Fujieda K. OTX2 loss of function mutation causes anophthalmia and combined pituitary hormone deficiency with a small anterior and ectopic posterior pituitary. J Clin Endocrinol Metab.2009;94(1):314-319.
  30. Dattani MT, Martinez-Barbera JP, Thomas PQ, Brickman JM, Gupta R, Martensson IL, Toresson H, Fox M, Wales JK, Hindmarsh PC, Krauss S, Beddington RS, Robinson IC. Mutations in the homeobox gene HESX1/Hesx1 associated with septo-optic dysplasia in human and mouse. Nat Genet.1998;19(2):125-133.
  31. Durmaz B, Cogulu O, Dizdarer C, Stobbe H, Pfaeffle R, Ozkinay F. A novel homozygous HESX1 mutation causes panhypopituitarism without midline defects and optic nerve anomalies J Pediatr Endocr Met.2011;24(9-10):779-782.
  32. Thomas PQ, Dattani MT, Brickman JM, McNay D, Warne G, Zacharin M, Cameron F, Hurst J, Woods K, Dunger D, Stanhope R, Forrest S, Robinson IC, Beddington RS. Heterozygous HESX1 mutations associated with isolated congenital pituitary hypoplasia and septo-optic dysplasia. Hum Mol Genet.2001;10(1):39-45.
  33. Newbern K, Natrajan N, Kim HG, Chorich LP, Halvorson LM, Cameron RS, Layman LC. Identification of HESX1 mutations in Kallmann syndrome. Fertil Steril.2013;99(7):1831-1837.
  34. Williamson KA, Yates TM, FitzPatrick DR. SOX2 Disorder. In: Adam MP, Everman DB, Mirzaa GM, Pagon RA, Wallace SE, Bean LJH, Gripp KW, Amemiya A, eds. GeneReviews((R)). Seattle (WA)1993.
  35. Fauquier T, Rizzoti K, Dattani M, Lovell-Badge R, Robinson ICAF. SOX2-expressing progenitor cells generate all of the major cell types in the adult mouse pituitary gland. Proc Natl Acad Sci U S A 2008;105(8):2907-2912.
  36. Williamson KA, Hever AM, Rainger J, Rogers RC, Magee A, Fiedler Z, Keng WT, Sharkey FH, McGill N, Hill CJ, Schneider A, Messina M, Turnpenny PD, Fantes JA, van Heyningen V, FitzPatrick DR. Mutations in SOX2 cause anophthalmia-esophageal-genital (AEG) syndrome. Hum Mol Genet.2006;15(9):1413-1422.
  37. Kelberman D, Rizzoti K, Avilion A, Bitner-Glindzicz M, Cianfarani S, Collins J, Chong WK, Kirk JMW, Achermann JC, Ross R, Carmignac D, Lovell-Badge R, Robinson ICAF, Dattani MT. Mutations within SOX2/SOX2 are associated with abnormalities in the hypothalamo-pituitary-gonadal axis in mice and humans. . The Journal of Clinical Investigation 2006;116(9):2442-2455.
  38. Chassaing N, A AC, A VV, A A, J-L JA, O OB-T, O B-B, H D, B D-B, B G-D, Giuliano F GM, M H-E, B I, M-L J, D L, D M-C, M M-D, S O, O P, L P, C Q, S S, A T, C T-R, J K, P C. Molecular findings and clinical data in a cohort of 150 patients with anophthalmia/ microphthalmia. Clin Genet.2013.
  39. Blum WF, Klammt J, Amselem S, Pfaffle HM, Legendre M, Sobrier ML, Luton MP, Child CJ, Jones C, Zimmermann AG, Quigley CA, Cutler GB, Jr., Deal CL, Lebl J, Rosenfeld RG, Parks JS, Pfaffle RW. Screening a large pediatric cohort with GH deficiency for mutations in genes regulating pituitary development and GH secretion: Frequencies, phenotypes and growth outcomes. EBioMedicine.2018;36:390-400.
  40. Laumonnier F, Ronce N, Hamel BCJ, Thomas P, Lespinasse J, Raynaud M, Paringaux C, Bokhoven Hv, Kalscheuer V, Fryns J-P, Chelly J, Moraine C, Briault S. Transcription factor SOX3 is involved in X-linked mental retardation with growth hormone deficiency. Am J Hum Genet 2002;71(6):1450-1455.
  41. Gregory LC, Gergics P, Nakaguma M, Bando H, Patti G, McCabe MJ, Fang Q, Ma Q, Ozel AB, Li JZ, Poina MM, Jorge AAL, Benedetti AFF, Lerario AM, Arnhold IJP, Mendonca BB, Maghnie M, Camper SA, Carvalho LRS, Dattani MT. The phenotypic spectrum associated with OTX2 mutations in humans. Eur J Endocrinol.2021;185(1):121-135.
  42. Mortensen AH, Schade V, Lamonerie T, Camper SA. Deletion of OTX2 in neural ectoderm delays anterior pituitary development. Hum Mol Genet.2015;24(4):939-953.
  43. Cunha D, Arno G, Corton M, Moosajee M. The Spectrum of PAX6 Mutations and Genotype-Phenotype Correlations in the Eye. Genes.2019;10:1050-1072.
  44. Kioussi C, O’Connell S, St-Onge L, Treier M, Gleiberman AS, Gruss P, Rosenfeld MG. <i>Pax6</i> is essential for establishing ventral-dorsal cell boundaries in pituitary gland development. Proceedings of the National Academy of Sciences.1999;96(25):14378-14382.
  45. Hergott-Faure L, Borot S, Kleinclauss C, Abitbol M, Penfornis A. Pituitary function and glucose tolerance in a family with a PAX6 mutation. Ann Endocrinol (Paris).2012;73(6):510-514.
  46. Takagi M, Nagasaki K, Fujiwara I, Ishii T, Amano N, Asakura Y, Muroya K, Hasegawa Y, Adachi M, Hasegawa T. Heterozygous defects in PAX6 gene and congenital hypopituitarism. Eur J Endocrinol.2015;172(1):37-45.
  47. Shimo N, Yasuda T, Kitamura T, Matsushita K, Osawa S, Yamamoto Y, Kozawa J, Otsuki M, Funahashi T, Imagawa A, Kaneto H, Nishida K, Shimomura I. Aniridia with a heterozygous PAX6 mutation in which the pituitary function was partially impaired. Intern Med.2014;53(1):39-42.
  48. Brachet C, Kozhemyakina EA, Boros E, Heinrichs C, Balikova I, Soblet J, Smits G, Vilain C, Mathers PH. Truncating RAX Mutations: Anophthalmia, Hypopituitarism, Diabetes Insipidus, and Cleft Palate in Mice and Men. J Clin Endocrinol Metab.2019;104(7):2925-2930.
  49. Gaston-Massuet C, McCabe MJ, Scagliotti V, Young RM, Carreno G, Gregory LC, Jayakody SA, Pozzi S, Gualtieri A, Basu B, Koniordou M, Wu C-I, Bancalari RE, Rahikkala E, Veijola R, Lopponen T, Graziola F, Turton J, Signore M, Gharavy SNM, Charolidi N, Sokol SY, Andoniadou CL, Wilson SW, Merrill BJ, Dattani MT, Martinez-Barberaa JP. Transcription factor 7-like 1 is involved in hypothalamo–pituitary axis development in mice and humans. PNAS.2016;113(5):E548-E557.
  50. De Rienzo F, Mellone S, Bellone S, Babu D, Fusco I, Prodam F, Petri A, Muniswamy R, De Luca F, Salerno M, Momigliano-Richardi P, Bona G, Giordano M, Italian Study Group on Genetics of C. Frequency of genetic defects in combined pituitary hormone deficiency: a systematic review and analysis of a multicentre Italian cohort. Clin Endocrinol (Oxf).2015;83(6):849-860.
  51. Rajab A, Kelberman D, de Castro SC, Biebermann H, Shaikh H, Pearce K, Hall CM, Shaikh G, Gerrelli D, Grueters A, Krude H, Dattani MT. Novel mutations in LHX3 are associated with hypopituitarism and sensorineural hearing loss. Hum Mol Genet.2008;17(14):2150-2159.
  52. Ellsworth BS, Butts DL, Camper SA. Mechanisms underlying pituitary hypoplasia and failed cell specification in Lhx3-deficient mice. Dev Biol.2008;313(1):118-129.
  53. Xatzipsalti M, Voutetakis A, Stamoyannou L, Chrousos GP, Kanaka-Gantenbein C. Congenital Hypopituitarism: Various Genes, Various Phenotypes. Horm Metab Res.2019;51:81-90.
  54. Bechtold-Dalla Pozza S, Hiedl S, Roeb J, Lohse P, Malik RE, Park S, Duran-Prado M, Rhodes SJ. A recessive mutation resulting in a disabling amino acid substitution (T194R) in the LHX3 homeodomain causes combined pituitary hormone deficiency. Horm Res Paediatr.2012;77(1):41-51.
  55. Sobrier ML, Brachet C, Vie-Luton MP, Perez C, Copin B, Legendre M, Heinrichs C, Amselem S. Symptomatic heterozygotes and prenatal diagnoses in a nonconsanguineous family with syndromic combined pituitary hormone deficiency resulting from two novel LHX3 mutations. J Clin Endocrinol Metab.2012;97(3):E503-509.
  56. Cohen E, Maghnie M, Collot N, Leger J, Dastot F, Polak M, Rose S, Touraine P, Duquesnoy P, Tauber M, Copin B, Bertrand AM, Brioude F, Larizza D, Edouard T, Gonzalez Briceno L, Netchine I, Oliver-Petit I, Sobrier ML, Amselem S, Legendre M. Contribution of LHX4 Mutations to Pituitary Deficits in a Cohort of 417 Unrelated Patients. J Clin Endocrinol Metab.2017;102(1):290-301.
  57. Machinis K, Amselem S. Functional Relationship between LHX4 and POU1F1 in Light of the LHX4 Mutation Identified in Patients with Pituitary Defects. J Clin Endocrinol Metab.2005;90(9):5456-5462.
  58. Liu Y, Fan M, Yu S, Zhou Y, Wang J, Yuan J, Qiang B. cDNA cloning, chromosomal localization and expression pattern analysis of human LIM-homeobox gene LHX4. Brain Res.2002;928(1-2):147-155.
  59. Colvin SC, Mullen RD, Pfaeffle RW, Rhodes SJ. LHX3 and LHX4 transcription factors in pituitary development and disease. Pediatr Endocrinol Rev.2009;6 Suppl 2:283-290.
  60. Gregory LC, Humayun KN, Turton JP, McCabe MJ, Rhodes SJ, Dattani MT. Novel Lethal Form of Congenital Hypopituitarism Associated With the First Recessive LHX4 Mutation. J Clin Endocrinol Metab.2015;100(6):2158-2164.
  61. Tajima T, Hattori T, Nakajima T, Okuhara K, Tsubaki J, Fujieda K. A novel missense mutation (P366T) of the LHX4 gene causes severe combined pituitary hormone deficiency with pituitary hypoplasia, ectopic posterior lobe and a poorly developed sella turcica. Endocr J.2007;54(4):637-641.
  62. Filges I, Bischof-Renner A, Rothlisberger B, Potthoff C, Glanzmann R, Gunthard J, Schneider J, Huber AR, Zumsteg U, Miny P, Szinnai G. Panhypopituitarism presenting as life-threatening heart failure caused by an inherited microdeletion in 1q25 including LHX4. Pediatrics.2012;129(2):e529-534.
  63. Franca MM, Jorge AA, Carvalho LR, Costalonga EF, Otto AP, Correa FA, Mendonca BB, Arnhold IJ. Relatively high frequency of non-synonymous GLI2 variants in patients with congenital hypopituitarism without holoprosencephaly. Clin Endocrinol (Oxf).2013;78(4):551-557.
  64. Wang Y, Martin JF, Bai CB. Direct and indirect requirements of Shh/Gli signaling in early pituitary development. Dev Biol.2010;348(2):199-209.
  65. Roessler E, Du YZ, Mullor JL, Casas E, Allen WP, Gillessen-Kaesbach G, Roeder ER, Ming JE, Ruiz i Altaba A, Muenke M. Loss-of-function mutations in the human GLI2 gene are associated with pituitary anomalies and holoprosencephaly-like features. Proc Natl Acad Sci U S A.2003;100(23):13424-13429.
  66. Bear K, Solomon BD, Antonini S, Arnhold IJP, França MM, Gerkes EH, Grange DK, Hadley DW, Jääskeläinen J, Paulo SS, Rump P, Stratakis CA, Thompson EM, Willis M, Winder TL, Jorge AAL, Roessler E, Muenke M. Pathogenic mutations in GLI2 cause a specific phenotype that is distinct from holoprosencephaly. J Med Genet.2014;51(6):413-418.
  67. Paulo S, Fernandes-Rosa FL, Turatti W, Coeli-Lacchini FB, Martinelli CE, Nakiri GS, Moreira AC, Santos AC, Castro Md, Antonini SR. Sonic Hedgehog mutations are not a common cause of congenital hypopituitarism in the absence of complex midline cerebral defects. Clinical Endocrinology.2015;82:562-569.
  68. Biesecker LG. GLI3-Related Pallister-Hall Syndrome. In: Adam MP, Everman DB, Mirzaa GM, Pagon RA, Wallace SE, Bean LJH, Gripp KW, Amemiya A, eds. GeneReviews((R)). Seattle (WA)1993.
  69. Haddad-Tóvolli R, Paul F, Zhang Y, Zhou X, Theil T, Puelles L, Blaess S, Alvarez-Bolado G. Differential requirements for Gli2 and Gli3 in the regional specification of the mouse hypothalamus. Front Neuroanat.2015;9.
  70. Fang Q, George A, Brinkmeier M, Mortensen A, Gergics P, Cheung L, Daly AZ, A Ajmal, Millán MIP, Ozel A, Kitzman J, Mills R, Li J, Camper S. Genetics of Combined Pituitary Hormone Deficiency: Roadmap into the Genome Era. Endocrine Reviews.2016;37(6):636-675.
  71. Hall JG, Pallister PD, Clarren SK, Beckwith JB, Wiglesworth FW, Fraser FC, Cho S, Benke PJ, Reed SD. Congenital hypothalamic hamartoblastoma, hypopituitarism, imperforate anus and postaxial polydactyly--a new syndrome? Part I: clinical, causal, and pathogenetic considerations. Am J Med Genet.1980;7(1):47-74.
  72. Biesecker L, Graham J. Pallister-Hall Syndrome. J Med Genet.1996;33:585-589.
  73. Dubourg C, Bendavid C, Pasquier L, Henry C, Odent S, David V. Holoprosencephaly. Orphanet J Rare Dis.2007;2(8):8.
  74. Hahn JS, Hahn SM, Kammann H, Barkovich AJ, Clegg NJ, Delgado MR, Levey E. Endocrine disorders associated with holoprosencephaly. J Pediatr Endocrinol Metab.2005;18(10):935-941.
  75. Dubourg C, Kim A, Watrin E, de Tayrac M, Odent S, David V, Dupe V. Recent advances in understanding inheritance of holoprosencephaly. Am J Med Genet C Semin Med Genet.2018;178(2):258-269.
  76. Tekendo-Ngongang C, Muenke M, Kruszka P. Holoprosencephaly Overview. In: Adam MP, Everman DB, Mirzaa GM, Pagon RA, Wallace SE, Bean LJH, Gripp KW, Amemiya A, eds. GeneReviews((R)). Seattle (WA)1993.
  77. Tasdemir S, Sahin I, Cayir A, Doneray H, Solomon BD, Muenke M, Yuce I, Tatar A. Holoprosencephaly: ZIC2 mutation in a case with panhypopituitarism. J Pediatr Endocrinol Metab.2014;27(7-8):777-781.
  78. Bando H, Brinkmeier M, Gergics P, Fang Q, Mortensen AH, Ozel AB, Ma Q, Li J, Reynaud R, Castinetti F, Brue TC, Camper SA. SIX3 Variant Causes Pituitary Stalk Interruption Syndrome and Combined Pituitary Hormone Deficiency. J Endocrine Soc.2021;5(1):A530.
  79. Tatsi C, Sertedaki A, Voutetakis A, Valavani E, Magiakou M-A, Kanaka-Gantenbein C, Chrousos GP, Dacou-Voutetakis C. Pituitary Stalk Interruption Syndrome and Isolated Pituitary Hypoplasia May Be Caused by Mutations in Holoprosencephaly-Related Genes. J Clin Endocrinol Metab.2013;98(4):779-784.
  80. Obara‑Moszyńska M, Budny B, Kałużna M, Zawadzka K, Jamsheer A, Rohde A, Ruchała M, Ziemnicka K, Niedziela M. CDON gene contributes to pituitary stalk interruption syndrome associated with unilateral facial and abducens nerve palsy. Journal of Applied Genetics 2021;62:621-629.
  81. Karaca E, Buyukkaya R, Pehlivan D, Charng WL, Yaykasli KO, Bayram Y, Gambin T, Withers M, Atik MM, Arslanoglu I, Bolu S, Erdin S, Buyukkaya A, Yaykasli E, Jhangiani SN, Muzny DM, Gibbs RA, Lupski JR. Whole-exome sequencing identifies homozygous GPR161 mutation in a family with pituitary stalk interruption syndrome. J Clin Endocrinol Metab.2015;100(1):E140-147.
  82. Bashamboo A, Bignon-Topalovic J, Rouba H, McElreavey K, Brauner R. A Nonsense Mutation in the Hedgehog Receptor CDON Associated With Pituitary Stalk Interruption Syndrome. J Clin Endocrinol Metab.2016;101(1):12-15.
  83. Wang CZ, Guo LL, Han BY, Su X, Guo QH, Mu YM. Pituitary Stalk Interruption Syndrome: From Clinical Findings to Pathogenesis. J Neuroendocrinol.2017;29(1).
  84. Bashamboo A, Bignon-Topalovic J, Moussi N, McElreavey K, Brauner R. Mutations in the Human ROBO1 Gene in Pituitary Stalk Interruption Syndrome. J Clin Endocrinol Metab.2017;102(7):2401-2406.
  85. Dateki S, Watanabe S, Mishima H, Shirakawa T, Morikawa M, Kinoshita E, Yoshiura KI, Moriuchi H. A homozygous splice site ROBO1 mutation in a patient with a novel syndrome with combined pituitary hormone deficiency. J Hum Genet.2019;64(4):341-346.
  86. Vajravelu ME, Chai J, Krock B, Baker S, Langdon D, Alter C, De Leon DD. Congenital Hyperinsulinism and Hypopituitarism Attributable to a Mutation in FOXA2. J Clin Endocrinol Metab.2018;103(3):1042-1047.
  87. Dayem-Quere M, Giuliano F, Wagner-Mahler K, Massol C, Crouzet-Ozenda L, Lambert JC, Karmous-Benailly H. Delineation of a region responsible for panhypopituitarism in 20p11.2. Am J Med Genet A.2013;161A(7):1547-1554.
  88. Williams PG, Wetherbee JJ, Rosenfeld JA, Hersh JH. 20p11 deletion in a female child with panhypopituitarism, cleft lip and palate, dysmorphic facial features, global developmental delay and seizure disorder. Am J Med Genet A.2011;155A(1):186-191.
  89. Garcia-Heras J, Kilani RA, Martin RA, Lamp S. A deletion of proximal 20p inherited from a normal mosaic carrier mother in a newborn with panhypopituitarism and craniofacial dysmorphism. Clin Dysmorphol.2005;14(3):137-140.
  90. Giri D, Vignola ML, Gualtieri A, Scagliotti V, McNamara P, Peak M, Didi M, Gaston-Massuet C, Senniappan S. Novel FOXA2 mutation causes Hyperinsulinism, Hypopituitarism with Craniofacial and Endoderm-derived organ abnormalities. Hum Mol Genet.2017;26(22):4315-4326.
  91. Heddad Masson M, Poisson C, Guerardel A, Mamin A, Philippe J, Gosmain Y. Foxa1 and Foxa2 regulate alpha-cell differentiation, glucagon biosynthesis, and secretion. Endocrinology.2014;155(10):3781-3792.
  92. Tsai EA, Grochowski CM, Falsey AM, Rajagopalan R, Wendel D, Devoto M, Krantz ID, Loomes KM, Spinner NB. Heterozygous deletion of FOXA2 segregates with disease in a family with heterotaxy, panhypopituitarism, and biliary atresia. Hum Mutat.2015;36(6):631-637.
  93. Stekelenburg C, Gerster K, Blouin JL, Lang-Muritano M, Guipponi M, Santoni F, Schwitzgebel VM. Exome sequencing identifies a de novo FOXA2 variant in a patient with syndromic diabetes. Pediatr Diabetes.2019;20(3):366-369.
  94. Gregory LC, Ferreira CB, Young-Baird SK, Williams HJ, Harakalova M, Haaften Gv, Rahman SA, Gaston-Massuet C, Kelberman D, Qasim W, Camper SA, Dever TE, Shah P, Robinson ICAF, Dattani MT. Impaired EIF2S3 function associated with a novel phenotype of X-linked hypopituitarism with glucose dysregulatio. EBioMedicine 2019;42:470-480.
  95. Skopkova M, Hennig F, Shin BS, Turner CE, Stanikova D, Brennerova K, Stanik J, Fischer U, Henden L, Muller U, Steinberger D, Leshinsky-Silver E, Bottani A, Kurdiova T, Ukropec J, Nyitrayova O, Kolnikova M, Klimes I, Borck G, Bahlo M, Haas SA, Kim JR, Lotspeich-Cole LE, Gasperikova D, Dever TE, Kalscheuer VM. EIF2S3 Mutations Associated with Severe X-Linked Intellectual Disability Syndrome MEHMO. Hum Mutat.2017;38(4):409-425.
  96. Takuma N, Sheng HZ, Furuta Y, Ward JM, Sharma K, Hogan BL, Pfaff SL, Westphal H, Kimura S, Mahon KA. Formation of Rathke's pouch requires dual induction from the diencephalon. Development.1998;125(23):4835-4840.
  97. Rodriguez-Contreras FJ, Marban-Calzon M, Vallespin E, Del Pozo A, Solis-Lopez M, Lobato-Vidal N, Fernandez-Elvira M, Del Valle Rex-Romero M, Heath KE, Gonzalez-Casado I, Campos-Barros A. Loss of function BMP4 mutation supports the implication of the BMP/TGF-beta pathway in the etiology of combined pituitary hormone deficiency. Am J Med Genet A.2019;179(8):1591-1597.
  98. Johanna Hietamäki, Gregory LC, Ayoub S, Iivonen A-P, Vaaralahti K, Liu X, Brandstack N, Buckton AJ, Laine T, Känsäkoski J, Hero M, Miettinen PJ, Varjosalo M, Wakeling E, Dattani MT, Raivio T. Loss-of-Function Variants in TBC1D32 Underlie Syndromic Hypopituitarism. J Clin Endocrinol Metab.2020;105(6):1748-1758.
  99. Webb EA, AlMutair A, Kelberman D, Bacchelli C, Chanudet E, Lescai F, Andoniadou CL, Banyan A, Alsawaid A, Alrifai MT, Alahmesh MA, Balwi M, Mousavy-Gharavy SN, Lukovic B, Burke D, McCabe MJ, Kasia T, Kleta R, Stupka E, Beales PL, Thompson DA, Chong WK, Alkuraya FS, Martinez-Barbera JP, Sowden JC, Dattani MT. ARNT2 mutation causes hypopituitarism, post-natal microcephaly, visual and renal anomalies. Brain.2013;136(Pt 10):3096-3105.
  100. Jadhav S, Diwaker C, Lila A, Gada J, Kale S, Sarathi V, Thadani P, Arya S, Patil V, Shah N, Bandgar T, Ajmal A. POU1F1 mutations in combined pituitary hormone deficiency: differing spectrum of mutations in a Western‑Indian cohort and systematic analysis of world literature. Pituitary.2021;24:657-669.
  101. McLennan K, Jeske Y, Cotterill A, Cowley D, Penfold J, Jones T, Howard N, Thomsett M, Choong C. Combined pituitary hormone deficiency in Australian children: clinical and genetic correlates. Clin Endocrinol (Oxf).2003;58(6):785-794.
  102. Anderson B, Rosenfeld M. Pit-1 Determines Cell types during Development of the Anterior Pituitary Gland The Journal of Biological Chemistry.1994;269(47):29335-29338.
  103. Tatsumi K, Miyai K, Notomi T, Kaibe K, Amino N, Mizuno Y, Kohno H. Cretinism with combined hormone deficiency caused by a mutation in the PIT1 gene. Nat Genet.1992;1(1):56-58.
  104. Ward RD, Raetzman LT, Suh H, Stone BM, Nasonkin IO, Camper SA. Role of PROP1 in pituitary gland growth. Mol Endocrinol.2005;19(3):698-710.
  105. Bottner A, Keller E, Kratzsch J, Stobbe H, Weigel JF, Keller A, Hirsch W, Kiess W, Blum WF, Pfaffle RW. PROP1 mutations cause progressive deterioration of anterior pituitary function including adrenal insufficiency: a longitudinal analysis. J Clin Endocrinol Metab.2004;89(10):5256-5265.
  106. Mullis PE, Robinson ICAF, Salemi S, Eble A, Besson A, Vuissoz J-M, Deladoey J, Simon D, Czernichow P, Binder G. Isolated Autosomal Dominant Growth Hormone Deficiency: An Evolving Pituitary Deficit? A Multicentre Follow-Up Study. J Clin Endocrinol Metab.2005;90(4):2089-2096.
  107. Cerbone M, Dattani MT. Progression from isolated growth hormone deficiency to combined pituitary hormone deficiency. Growth Horm IGF Res.2017;37:19-25.
  108. Mullis PE. Genetics of isolated growth hormone deficiency. J Clin Res Pediatr Endocrinol.2010;2(2):52-62.
  109. Alatzoglou KS, Turton JP, Kelberman D, Clayton PE, Mehta A, Buchanan C, Aylwin S, Crowne EC, Christesen HT, Hertel NT, Trainer PJ, Savage MO, Raza J, Banerjee K, Sinha SK, Ten S, Mushtaq T, Brauner R, Cheetham TD, Hindmarsh PC, Mullis PE, Dattani MT. Expanding the spectrum of mutations in GH1 and GHRHR: genetic screening in a large cohort of patients with congenital isolated growth hormone deficiency. J Clin Endocrinol Metab.2009;94(9):3191-3199.
  110. Phillips JA, 3rd, Hjelle BL, Seeburg PH, Zachmann M. Molecular basis for familial isolated growth hormone deficiency. Proc Natl Acad Sci U S A.1981;78(10):6372-6375.
  111. Wajnrajch MP, Gertner JM, Harbison MD, Chua SC, Jr., Leibel RL. Nonsense mutation in the human growth hormone-releasing hormone receptor causes growth failure analogous to the little (lit) mouse. Nat Genet.1996;12(1):88-90.
  112. Mazhar M, Waseem M. Agammaglobulinaemia. StatPearls [Internet].2022.
  113. Child CJ, Blum WF, Deal C, Zimmermann AG, Quigley CA, Drop SLS, Cutler GB, Rosenfeld RG. Development of additional pituitary hormone deficiencies in pediatric patients originally diagnosed with isolated growth hormone deficiency due to organic causes. Eur J Endocrinol.2016;174(5):669-679.
  114. Casteras A, Kratzsch J, Ferrandez A, Zafon C, Carrascosa A, Mesa J. Clinical challenges in the management of isolated GH deficiency type IA in adulthood. Endocrinol Diabetes Metab Case Rep.2014;2014:130057.
  115. Lanting CI, Tijn DAv, Loeber JG, Vulsma T, Vijlder JJMd, Verkerk PH. Clinical Effectiveness and Cost-Effectiveness of the Use of the Thyroxine/Thyroxine-Binding Globulin Ratio to Detect Congenital Hypothyroidism of Thyroidal and Central Origin in a Neonatal Screening Program. Pediatrics.2005;116(1):168-173.
  116. Fisher DA. Second International Conference on Neonatal Thyroid Screening: progress report. J Pediatr.1983;102(5):653-654.
  117. Zwaveling-Soonawala N, Naafs JC, Verkerk PH, van Trotsenburg ASP. Mortality in Children With Early-Detected Congenital Central Hypothyroidism. J Clin Endocrinol Metab.2018;103(8):3078-3082.
  118. Tajima T, Nakamura A, Oguma M, Yamazaki M. Recent advances in research on isolated congenital central hypothyroidism. Clin Pediatr Endocrinol.2019;28(3):69-79.
  119. Hayashizaki Y, Hiraoka Y, Endo Y, Miyai K, Matsubara K. Thyroid-stimulating hormone (TSH) deficiency caused by a single base substitution in the CAGYC region of the beta-subunit. EMBO J.1989;8(8):2291-2296.
  120. Couture C, Saveanu A, Barlier A, Carel JC, Fassnacht M, Fluck CE, Houang M, Maes M, Phan-Hug F, Enjalbert A, Drouin J, Brue T, Vallette S. Phenotypic homogeneity and genotypic variability in a large series of congenital isolated ACTH-deficiency patients with TPIT gene mutations. J Clin Endocrinol Metab.2012;97(3):E486-495.
  121. Stevens A, White A, eds. Adrenocorticotrophic Hormone. 6 ed. Philadelphia: Saunders Elsevier; 2010. Jameson JL, Groot LD, eds. Endocrinology; No. 1
  122. Derya-Kardelen-Al A, Poyrazoğlu Ş, Aslanger A, Yeşil G, Ceylaner S, Baş F, Darendeliler F. A Rare Cause of Adrenal Insufficiency – Isolated ACTH Deficiency Due to TBX19 Mutation: Long-Term Follow-Up of Two Cases and Review of the Literature. HORMONE RESEARCH IN PÆDIATRICS.2019;92:395-403.
  123. Weintrob N, Drouin J, Vallette-Kasic S, Taub E, Marom D, Lebenthal Y, Klinger G, Bron-Harlev E, Shohat M. Low Estriol Levels in the Maternal Triple-Marker Screen as a Predictor of Isolated AdrenocorticotropicHormone Deficiency Caused by a New Mutation in the TPIT Gene. Pediatrics.2006;117 (2):322-327.
  124. Stijnen P, Ramos-Molina B, O'Rahilly S, Creemers JW. PCSK1 Mutations and Human Endocrinopathies: From Obesity to Gastrointestinal Disorders. Endocr Rev.2016;37(4):347-371.
  125. Gregoric N, Groselj U, Bratina N, Debeljak M, Zerjav Tansek M, Suput Omladic J, Kovac J, Battelino T, Kotnik P, Avbelj Stefanija M. Two Cases With an Early Presented Proopiomelanocortin Deficiency-A Long-Term Follow-Up and Systematic Literature Review. Front Endocrinol (Lausanne).2021;12:689387.
  126. Mendiratta MS, Yang Y, Balazs AE, Willis AS, Eng CM, Karaviti LP, Potocki L. Early onset obesity and adrenal insufficiency associated with a homozygous POMC mutation. Int J Pediatr Endocrinol.2011;2011(1):5.
  127. Brue T, Quentien M, Khetchoumian K, Bensa M, Capo-Chichi J, Delemer B, Balsalobre A, Nassif C, Papadimitriou D, Pagnier A, Hasselmann C, Patry L, Schwartzentruber J, Souchon P, Takayasu S, Enjalbert A, Vliet GV, Majewski J, Drouin J, Samuels M. Mutations in NFKB2 and potential genetic heterogeneity in patients with DAVID syndrome, having variable endocrine and immune deficiencies. BMC Medical Genetics.2014;15(139).
  128. Lal RA, Bachrach LK, Hoffman AR, Inlora J, Rego S, Snyder MP, Lewis DB. A Case Report of Hypoglycemia and Hypogammaglobulinemia: DAVID Syndrome in a Patient With a Novel NFKB2 Mutation. J Clin Endocrinol Metab.2017;102(7):2127-2130.
  129. Young J, Xu C, Hietamaki J, Papadakis GE, Acierno JS, Maione L, Raivio T, Pitteloud N. Clinical Mangement of Congenital Hypogonadotrophic Hypogonadism. Endocrine Reviews.2019;40(2):669-710.
  130. Boehm U, Bouloux PM, Dattani MT, de Roux N, Dode C, Dunkel L, Dwyer AA, Giacobini P, Hardelin JP, Juul A, Maghnie M, Pitteloud N, Prevot V, Raivio T, Tena-Sempere M, Quinton R, Young J. Expert consensus document: European Consensus Statement on congenital hypogonadotropic hypogonadism--pathogenesis, diagnosis and treatment. Nat Rev Endocrinol.2015;11(9):547-564.
  131. Dijk DR, Bocca G, van Ravenswaaij-Arts CM. Growth in CHARGE syndrome: optimizing care with a multidisciplinary approach. J Multidiscip Healthc.2019;12:607-620.
  132. Pagon RA, Graham JM, Jr., Zonana J, Yong SL. Coloboma, congenital heart disease, and choanal atresia with multiple anomalies: CHARGE association. J Pediatr.1981;99(2):223-227.
  133. Aramaki M, Udaka T, Kosaki R, Makita Y, Okamoto N, Yoshihashi H, Oki H, Nanao K, Moriyama N, Oku S, Hasegawa T, Takahashi T, Fukushima Y, Kawame H, Kosaki K. Phenotypic spectrum of CHARGE syndrome with CHD7 mutations. J Pediatr.2006;148(3):410-414.
  134. Asakura Y, Toyota Y, Muroya K, Kurosawa K, Fujita K, Aida N, Kawame H, Kosaki K, Adachi M. Endocrine and radiological studies in patients with molecularly confirmed CHARGE syndrome. J Clin Endocrinol Metab.2008;93(3):920-924.
  135. Pinto G, Abadie V, Mesnage R, Blustajn J, Cabrol S, Amiel J, Hertz-Pannier L, Bertrand AM, Lyonnet S, Rappaport R, Netchine I. CHARGE syndrome includes hypogonadotropic hypogonadism and abnormal olfactory bulb development. J Clin Endocrinol Metab.2005;90(10):5621-5626.
  136. Shoji Y, Ida S, Etani Y, Yamada H, Kayatani F, Suzuki Y, Kosaki K, Okamoto N. Endocrinological Characteristics of 25 Japanese Patients with CHARGE Syndrome. Clin Pediatr Endocrinol.2014;23(2):45-51.
  137. Sarfati J, Fouveaut C, Leroy C, Jeanpierre M, Hardelin J-P, Dode C. Greater prevalence of PROKR2 mutations in Kallmann syndrome patients from the Maghreb than in European patients. Eur J Endocrinol.2013;169:805-809.
  138. Ericson J, Norlin S, Jessell TM, Edlund T. Integrated FGF and BMP signaling controls the progression of progenitor cell differentiation and the emergence of pattern in the embryonic anterior pituitary. Development.1198;125:1005-1015.
  139. Davis SW, Ellsworth BS, Perez Millan MI, Gergics P, Schade V, Foyouzi N, Brinkmeier ML, Mortensen AH, Camper SA. Pituitary gland development and disease: from stem cell to hormone production. Curr Top Dev Biol.2013;106:1-47.
  140. Tsai PS, Moenter SM, Postigo HR, El Majdoubi M, Pak TR, Gill JC, Paruthiyil S, Werner S, Weiner RI. Targeted expression of a dominant-negative fibroblast growth factor (FGF) receptor in gonadotropin-releasing hormone (GnRH) neurons reduces FGF responsiveness and the size of GnRH neuronal population. Mol Endocrinol.2005;19(1):225-236.
  141. Erbas IM, Paketci A, Acar S, Kotan LD, Demir K, Abaci A, Bober E. A nonsense variant in FGFR1: a rare cause of combined pituitary hormone deficiency. J Pediatr Endocrinol Metab.2020;33(12):1613-1615.
  142. Fukami M, Iso M, Sato N, Igarashi M, Seo M, Kazukawa I, Kinoshita E, Dateki S, Ogata T. Submicroscopic deletion involving the fibroblast growth factor receptor 1 gene in a patient with combined pituitary hormone deficiency. Endocr J.2013;60(8):1013-1020.
  143. Di Iorgi N, Napoli F, Allegri AE, Olivieri I, Bertelli E, Gallizia A, Rossi A, Maghnie M. Diabetes insipidus--diagnosis and management. Horm Res Paediatr.2012;77(2):69-84.
  144. Schernthaner-Reiter MH, Stratakis CA, Luger A. Genetics of Diabetes Insipidus. Endocrinol Metab Clin North Am.2017;46(2):305-334.
  145. Boutzios G, Livadas S, Marinakis E, Opie N, Economou F, Diamanti-Kandarakis E. Endocrine and metabolic aspects of the Wolfram syndrome. Endocrine.2011;40(1):10-13.
  146. Perrotta S, Di Iorgi N, Della Ragione F, Scianguetta S, Borriello A, Allegri AEM, Ferraro M, Santoro C, Napoli F, Calcagno A, Giaccardi M, Cappa M, Salerno MC, Cozzolino D, Maghnie M. Early-onset central diabetes insipidus is associated with de novo arginine vasopressin-neurophysin II or Wolfram syndrome 1 gene mutations. European Journal of Endocrinology.2015 172(4):461-472.
  147. Heksch R, Kamboj M, Anglin K, Obrynba K. Review of Prader-Willi syndrome: the endocrine approach. Transl Pediatr.2017;6(4):274-285.
  148. Miller JL, Goldstone AP, Couch JA, Shuster J, He G, Driscoll DJ, Liu Y, Schmalfuss IM. Pituitary abnormalities in Prader-Willi syndrome and early onset morbid obesity. Am J Med Genet A.2008;146A(5):570-577.
  149. Tauber M, Barbeau C, Jouret B, Pienkowski C, Malzac P, Moncla A, Rochiccioli P. Auxological and endocrine evolution of 28 children with Prader-Willi syndrome: effect of GH therapy in 14 children. Horm Res.2000;53(6):279-287.
  150. Carrel AL, Myers SE, Whitman BY, Allen DB. Benefits of long-term GH therapy in Prader-Willi syndrome: a 4-year study. J Clin Endocrinol Metab.2002;87(4):1581-1585.
  151. Festen DA, de Lind van Wijngaarden R, van Eekelen M, Otten BJ, Wit JM, Duivenvoorden HJ, Hokken-Koelega AC. Randomized controlled GH trial: effects on anthropometry, body composition and body proportions in a large group of children with Prader-Willi syndrome. Clin Endocrinol (Oxf).2008;69(3):443-451.
  152. Haqq AM, Stadler DD, Jackson RH, Rosenfeld RG, Purnell JQ, LaFranchi SH. Effects of growth hormone on pulmonary function, sleep quality, behavior, cognition, growth velocity, body composition, and resting energy expenditure in Prader-Willi syndrome. J Clin Endocrinol Metab.2003;88(5):2206-2212.
  153. Myers SE, Whitman BY, Carrel AL, Moerchen V, Bekx MT, Allen DB. Two years of growth hormone therapy in young children with Prader-Willi syndrome: physical and neurodevelopmental benefits. Am J Med Genet A.2007;143A(5):443-448.
  154. Radicioni AF, Di Giorgio G, Grugni G, Cuttini M, Losacco V, Anzuini A, Spera S, Marzano C, Lenzi A, Cappa M, Crino A. Multiple forms of hypogonadism of central, peripheral or combined origin in males with Prader-Willi syndrome. Clin Endocrinol (Oxf).2012;76(1):72-77.
  155. Gregory LC, Shah P, Sanner JRF, Arancibia M, Hurst J, Jones WD, Spoudeas H, Le Quesne Stabej P, Williams HJ, Ocaka LA, Loureiro C, Martinez-Aguayo A, Dattani MT. Mutations in MAGEL2 and L1CAM Are Associated With Congenital Hypopituitarism and Arthrogryposis. J Clin Endocrinol Metab.2019;104(12):5737-5750.
  156. F. Castinetti, Brue. T, Deficiency. CPH, Cohen IL, eds. Growth Hormone Deficiency: Physiology and Clinical Management Boston Children's Hospital: Springer International; 2016.
  157. Suh H, Gage PJ, Drouin J, Camper SA. Pitx2 is required at multiple stages of pituitary organogenesis: pituitary primordium formation and cell specification. Development.2002;129(2):329-337.
  158. Liu S, Wang Z, Jiang J, Luo X, Hong Q, Zhang Y, OuYang H, Wei S, Liang J, Chen N, Zeng W. Severe forms of Johanson-Blizzard syndrome caused by two novel compound heterozygous variants in UBR1: Clinical manifestations, imaging findings and molecular genetics. Pancreatology.2020;20(3):562-568.
  159. Almashraki N, Abdulnabee MZ, Sukalo M, Alrajoudi A, Sharafadeen I, Zenker M. Johanson-Blizzard syndrome. World J Gastroenterol.2011;17(37):4247-4250.
  160. Hoffman WH, Lee JR, Kovacs K, Chen H, Yaghmai F. Johanson-Blizzard syndrome: autopsy findings with special emphasis on hypopituitarism and review of the literature. Pediatr Dev Pathol.2007;10(1):55-60.
  161. Hufnagel RB, Arno G, Hein ND, Hersheson J, Prasad M, Anderson Y, Krueger LA, Gregory LC, Stoetzel C, Jaworek TJ, Hull S, Li A, Plagnol V, Willen CM, Morgan TM, Prows CA, Hegde RS, Riazuddin S, Grabowski GA, Richardson RJ, Dieterich K, Huang T, Revesz T, Martinez-Barbera JP, Sisk RA, Jefferies C, Houlden H, Dattani MT, Fink JK, Dollfus H, Moore AT, Ahmed ZM. Neuropathy target esterase impairments cause Oliver-McFarlane and Laurence-Moon syndromes. J Med Genet.2015;52(2):85-94.
  162. Lisbjerg K, Andersen MKG, Bertelsen M, Brost AG, Buchvald FF, Jensen RB, Bisgaard AM, Rosenberg T, Tumer Z, Kessel L. Oliver McFarlane syndrome: two new cases and a review of the literature. Ophthalmic Genet.2021;42(4):464-473.
  163. Baer S, Afenja A, Smol T, Piton A, Gérard B, Alembik Y, Bienvenu T, Boursier G, Boute O, Colson C, Cordier M-P, Cormier-Daire V, Delobel B, Doco-Fenzy M, Duban-Bedu B, Fradin M, Geneviève D, Goldenberg A, Grelet M, Haye D, Heron D, Isidor B, Keren B, Lacombe D, Lèbre A-S, Lesca G, Masurel A, Mathieu-Dramard M, Nava C, Pasquier L, Petit A, Philip N, Piard J, Sukno S, Thevenon J, Van-Gils J, Vincent-Delorme C, Willems M, Schaefer E, Morin G. Wiedemann-Steiner syndrome as a major cause of syndromic intellectual disability: A study of 33 French cases. Clinical Genetics.2018;94:141-152.
  164. Sheppard SE, Campbell IM, Harr MH, Gold N, Li D, Bjornsson HT, Cohen JS, Fahrner JA, Fatemi A, Harris JR, Nowak C, Stevens CA, Grand K, Au M, Graham JM, Jr., Sanchez-Lara PA, Campo MD, Jones MC, Abdul-Rahman O, Alkuraya FS, Bassetti JA, Bergstrom K, Bhoj E, Dugan S, Kaplan JD, Derar N, Gripp KW, Hauser N, Innes AM, Keena B, Kodra N, Miller R, Nelson B, Nowaczyk MJ, Rahbeeni Z, Ben-Shachar S, Shieh JT, Slavotinek A, Sobering AK, Abbott MA, Allain DC, Amlie-Wolf L, Au PYB, Bedoukian E, Beek G, Barry J, Berg J, Bernstein JA, Cytrynbaum C, Chung BH, Donoghue S, Dorrani N, Eaton A, Flores-Daboub JA, Dubbs H, Felix CA, Fong CT, Fung JLF, Gangaram B, Goldstein A, Greenberg R, Ha TK, Hersh J, Izumi K, Kallish S, Kravets E, Kwok PY, Jobling RK, Knight Johnson AE, Kushner J, Lee BH, Levin B, Lindstrom K, Manickam K, Mardach R, McCormick E, McLeod DR, Mentch FD, Minks K, Muraresku C, Nelson SF, Porazzi P, Pichurin PN, Powell-Hamilton NN, Powis Z, Ritter A, Rogers C, Rohena L, Ronspies C, Schroeder A, Stark Z, Starr L, Stoler J, Suwannarat P, Velinov M, Weksberg R, Wilnai Y, Zadeh N, Zand DJ, Falk MJ, Hakonarson H, Zackai EH, Quintero-Rivera F. Expanding the genotypic and phenotypic spectrum in a diverse cohort of 104 individuals with Wiedemann-Steiner syndrome. Am J Med Genet A.2021;185(6):1649-1665.
  165. Adam MP, Hudgins L, Hannibal M. Kabuki Syndrome. In: Adam MP, Everman DB, Mirzaa GM, Pagon RA, Wallace SE, Bean LJH, Gripp KW, Amemiya A, eds. GeneReviews((R)). Seattle (WA)1993.
  166. Ito N, Ihara K, Miyake N, Hara T, Tsutsumi Y, Matsumoto N. Hypothalamic pituitary complications in Kabuki syndrome. Pituitary.2019;16:133-138.
  167. Dayal D, Giri D, Senniappan S. A rare association of central hypothyroidism and adrenal insufficiency in a boy with Williams-Beuren syndrome. Ann Pediatr Endocrinol Metab.2017;22(1):65-67.
  168. Levy-Shraga Y, Gothelf D, Pinchevski-Kadir S, Katz U, Modan-Moses D. Endocrine manifestations in children with Williams-Beuren syndrome. Acta Paediatr.2018;107(4):678-684.
  169. Güven A. Seven cases with Williams-Beuren syndrome: endocrine evaluation and long-term follow-up. J Pediatr Endocr Met.2016;30(2):159-165.
  170. Xekouki P, Fryssira H, Maniati-Christidi M, Amenta S, Karavitakis EM, Kanaka-Gantenbein C, Dacou-Voutetakis C. Growth hormone deficiency in a child with Williams-Beuren syndrome. The response to growth hormone therapy. J Pediatr Endocrinol Metab.2005;18(2):205-207.
  171. Garcia ML, Ty EB, Taban M, David Rothner A, Rogers D, Traboulsi EI. Systemic and ocular findings in 100 patients with optic nerve hypoplasia. J Child Neurol.2006;21(11):949-956.
  172. Bongsebandhu-Phubhakdi C, Tempark T, Supornsilchai V. Endocrine manifestations of PHACE syndrome. J Pediatr Endocrinol Metab.2019;32(8):797-802.
  173. Goddard DS, Liang MG, Chamlin SL, Svoren BM, Spack NP, Mulliken JB. Hypopituitarism in PHACES Association. Pediatr Dermatol.2006;23(5):476-480.
  174. Garzon MC, Epstein LG, Heyer GL, Frommelt PC, Orbach DB, Baylis AL, Blei F, Burrows PE, Chamlin SL, Chun RH, Hess CP, Joachim S, Johnson K, Kim W, Liang MG, Maheshwari M, McCoy GN, Metry DW, Monrad PA, Pope E, Powell J, Shwayder TA, Siegel DH, Tollefson MM, Vadivelu S, Lew SM, Frieden IJ, Drolet BA. PHACE Syndrome: Consensus-Derived Diagnosis and Care Recommendations. J Pediatr.2016;178:24-33 e22.