Archives

Bacterial Infections in Endocrinology

ABSTRACT

 

Bacteria are microscopic organisms that are ubiquitous in the environment and human body. Some bacteria exhibit symbiotic relationship with the human body, while other bacteria are harmful and cause various diseases. Bacteria may infect the endocrine glands either by direct invasion or local or hematogenous spread. Suppurative bacterial infections can involve the pituitary, thyroid, adrenals, and gonads. In the majority of cases, specific risk factors predispose the endocrine glands to such infections. This in turn may lead to temporary or permanent endocrine dysfunction. There may also be states of hormone excess following bacterial infections. This is particularly noted in cases of bacterial thyroiditis. Permanent endocrine dysfunction following bacterial infections will warrant life-long hormone replacement therapy. In acute stages of infection, intravenous or oral antibiotics are the cornerstone of management. The choice of antibiotic is guided by culture and sensitivity report. Sometimes, however, empirical antibiotic therapy may need to be continued as no organism may be isolated on culture. Empirical therapy should provide coverage for gram positive, gram negative, and anaerobic bacteria. If there is abscess formation in any endocrine gland, it may require aspiration and drainage. In this chapter, we have discussed the risk factors, bacteriology, clinical presentation, diagnosis, and management of common bacterial infections involving endocrine glands.

INTRODUCTION

 

The incidence of bacterial infections of endocrine glands is low when compared to that in other organs of the body. The endocrine glands that may be affected by bacterial infections are: pituitary, thyroid, adrenals and gonads. Bacterial infection of parathyroid glands is extremely rare. Certain risk factors may predispose the glands for infection.

 

In general, bacteria may be classified as gram positive, gram negative, and miscellaneous categories. The classification of medically important bacteria is highlighted in another chapter of the Endotext (1). Among all the bacteria, Mycobacterium tuberculosis remains the most common agent involving the endocrine glands (2). Mycobacterium tuberculosis is a weakly gram positive highly aerobic bacterium that can cause tuberculosis in any organ of the body. This organism can affect the adrenal glands and lead to primary adrenal insufficiency. In developing countries, tuberculosis remains the most common cause of primary adrenal insufficiency. Tuberculosis can also affect pituitary, thyroid, and gonads. In this chapter, we are discussing only adrenal tuberculosis, since tuberculosis of the Endocrine system has been covered in great details in another chapter (3). Apart from Mycobacterium tuberculosis, the other common bacteria that may affect the endocrine system are Staphylococcus aureus, Streptococcus pneumoniae, Neisseria meningitides, Escherichia coli, Chlamydia trachomatis, Pseudomonas aeruginosa, Klebsiella pneumoniae, Treponema pallidum, and Yersinia enterocolitica among others. We have tried to present a spectrum of bacterial infections of various endocrine glands including their clinical presentation, investigations, management, long-term prognosis, and follow up.

 

BACTERIAL INFECTIONS OF PITUITARY

 

Infections of the pituitary gland are rare but may cause clinical problems because of the non-specific nature of the presentation (4). Among the various infectious agents, bacterial infections including Mycobacterial infections seem to be the most common. The various bacterial agents causing infection of the pituitary gland are summarized in the table 1. The common bacterial infections of the pituitary gland are described below.

 

Table 1.  Bacterial Agents Causing Infection of Pituitary-Hypothalamus

Bacterial class

Organism

Gram positive bacteria

Staphylococcus aureus, Streptococcus pneumoniae

Gram negative bacteria

E coli, Pseudomonas aeruginosa, Neisseria meningitides

Spirochaete

Treponema pallidum

Mycobacterium

Mycobacterium tuberculosis

 

Pituitary Abscess

 

EPIDEMIOLOGY AND RISK FACTORS

 

Pituitary abscesses are a very rare clinical entity and account for less than 1% of pituitary lesions (4). The first case of pituitary surgery involving an abscess was described in 1848. Since then, there have been around 300 such cases reported in the literature (4, 5). Risk factors include underlying pituitary diseases such as a pituitary adenoma, Rathke’s cyst, craniopharyngioma, lymphocytic hypophysitis, immunocompromised states (uncontrolled diabetes mellitus, tuberculosis, HIV infection, after solid organ transplantation, chemotherapy, radiotherapy), history of surgical exploration in pituitary hypothalamic region, and spread of local infection from meninges and paranasal sinuses (5-7). Rarely, abscess may develop in a normal pituitary gland (6, 8). 

 

BACTERIOLOGY

 

In the majority of cases, culture is negative in pituitary abscess, with only 19.7% cases showing growth of bacteria (9).The most common organisms isolated are Streptococci and Staphylococci. Other bacterial organisms are Escherichia coli, Mycobacteria, Neisseria, and anaerobes (6, 10). As culture is negative in most of the cases, it is important for empirical antibiotic therapy to cover gram positive, gram negative and anaerobic bacteria. Rarely, a fungal etiology is seen.

 

CLINICAL PRESENTATION

 

Clinical presentation can be classified with respect to chronicity as: acute (within days to weeks), subacute (less than a month) and chronic (more than a month). Acute and subacute abscesses have fulminant presentation while chronic abscess has a more indolent course (5). In the initial stages, patients present with headache (67%), fever, meningismus, and malaise. With progression of the disease, neurological symptoms like altered sensorium, seizures, and coma can occur.

 

Extension of infection in nearby areas can lead to visual dysfunction (45%), extra ocular movement defects, and other cranial nerve palsies (4, 8, 9, 11).

 

Both anterior and posterior pituitary hormonal hypofunction can be seen with a pituitary abscess. In the largest series of pituitary abscesses with 60 cases over 23 years, anterior pituitary hormone deficiencies were reported in 81.8% patients whereas diabetes insipidus was reported in 47.9% of the patients. In the same study, 9.3% had isolated hypogonadism, 3.7% had isolated ACTH deficiency, 1.8% had isolated hypothyroidism, and 1.8% hypothyroidism and ACTH deficiency (9).

 

DIAGNOSIS

 

The investigation of choice for the diagnosis of pituitary abscess is MRI (Magnetic Resonance Imaging) with proper sellar cuts. On T1 weighted images, pituitary abscess appears iso-intense to hypo-intense while on T2 weighted images, it is iso-intense to hyper-intense. There is a characteristic rim of enhancement after gadolinium injection around the abscess site (9, 11). Diffusion-weighted imaging (DWI) shows high signal intensity with a decrease in the apparent diffusion coefficient in the region of pus collection (9, 11).

 

MANAGEMENT

 

Trans-nasal trans-sphenoidal surgery and drainage of the abscess is the treatment of choice. The sphenoid sinus may require exploration if extrasellar invasion is suspected. Along with surgical exploration, the patient should be started on intravenous antibiotics empirically with ceftriaxone (alternatives are cefotaxime and cefepime) along with metronidazole for anaerobic coverage. In case of suspicion for Staphylococcus aureus, vancomycin should be added (9, 12). Further intensification or alteration of antibiotics is subjected to clinical improvement and culture and sensitivity reports. Microbiological etiology may not be identified in the majority of cases. Hence, it is imperative to give proper broad-spectrum coverage empirically.

 

PROGNOSIS AND FOLLOW-UP

 

With current standard of care, mortality rate is 10 % and chance of recurrence is <13%. In about 25% of cases hormonal recovery occurs. After recovering from a pituitary abscess, these patients should be followed up by serial MRI at 3, 6 and 12 months (12). Monitoring for anterior and posterior pituitary hormone deficiency should be done in any patient with a pituitary abscess. Replacement with corticosteroid, thyroid, gonadal, and growth hormone therapy may be required if the patient develops deficiency of any of these hormones. Replacement with vasopressin therapy may be required if patient develops central diabetes insipidus following a pituitary abscess.

 

Hypopituitarism Caused by Treponema Pallidum Infection

 

Syphilis caused by Treponema pallidum (a spirochete) may involve the pituitary- hypothalamic region causing syphilitic gumma with non-caseating granulomas (13, 14). It is more common in patients with underlying human immune deficiency virus (HIV) infection. Diagnosis can be made by demonstration of the spirochete in the samples of sellar tissues following trans sphenoidal surgery. Immunological diagnosis can be made by measuring titers of anti-Treponemal antibody in the serum. Treatment consists of intravenous followed by oral antibiotics (13-15). Penicillin is the drug of choice for syphilis. In patients who are allergic to penicillin, doxycycline is a good alternative.

 

BACTERIAL INFECTIONS OF THE THYROID

 

It is rare for bacteria to invade the normal thyroid gland because of the rich vascular supply, good lymphatic drainage, separation of thyroid gland from other structures by fascial planes, high iodine content, and production of hydrogen peroxide inside the gland (16). Both iodine and hydrogen peroxide have bactericidal properties.

 

Acute Suppurative Thyroiditis

 

EPIDEMIOLOGY AND RISK FACTORS      

 

Acute suppurative thyroiditis is rare and is usually due to bacterial infection of the thyroid gland. In severe cases, it can lead to abscess formation and spread to surrounding structures leading to acute obstruction of the respiratory tract. More than 90% of the patients are less than 40 years of age, with females being more commonly affected than males (17, 18). The incidence of acute suppurative thyroiditis lies between 0.1% and 0.7% of all thyroidal illnesses(19). In children acute suppurative thyroiditis is usually due to persistent pyriform sinus and almost always affects the left lobe and is often recurrent (20-22).  Risk factors for acute suppurative thyroiditis are summarized in table 2 (23).

 

Table 2. Risk Factors for Acute Suppurative Thyroiditis

Common risk factors

Pyriform sinus fistula – more common in children and young adults and associated with recurrent disease

Immunocompromised status – AIDS, blood malignancies, uncontrolled diabetes (more common risk factor overall)

Other risk factors

Thyroglossal cyst

Patent foramen cecum

Congenital brachial fistula

Spread of adjacent suppurative infection into thyroid

Anterior esophageal perforation

Underlying thyroid disorders like chronic autoimmune thyroiditis, goiter, and thyroid malignancy

Fine need aspirations/biopsy of thyroid

Dental abscess/ treatment

Systemic autoimmune disorders

 

BACTERIOLOGY

 

Although bacterial agents account for the majority of cases, acute suppurative thyroiditis can also be caused by fungal (immunosuppressive status), parasitic, and tubercular etiology. Common bacterial organisms include Staphylococcus aureus, Streptococcus pyogenes, Staphylococcus epidermidis, and Streptococcus pneumoniae. Rarely other causative bacteria include Klebsiella species, Hemophilus influenzae, Streptococcus viridans, Arcanobacterium haemolyticum, Eikenella corrodensSalmonella species, and Enterobacteriaceae. In the context of immunosuppressed states like HIV-AIDS, acute suppurative thyroiditis can be caused by Mycobacterium tuberculosis, atypical mycobacteria, Salmonella species, Nocardia species and Treponema pallidum (19, 24).

 

CLINICAL PRESENTATION

 

Acute suppurative thyroiditis due to bacterial etiology has a very rapid onset and progression of symptoms if not addressed. The common manifestations are fever, neck pain, and dysphagia. Thyroid gland may be tender on palpation and sometimes there may be swelling with fluctuation suggestive of localized pus collection (25). Very rarely infection can spread to nearby anatomical structures resulting in a more dramatic presentation with stridor due to laryngeal involvement requiring urgent tracheostomy (26). It is important to differentiate this condition from subacute thyroiditis which also presents with systemic symptoms and neck pain (Table 3) (see below).

 

DIAGNOSIS AND MANAGEMENT

 

Laboratory investigations are consistent with acute inflammation characterized by leukocytosis with shift to left, elevated erythrocyte sedimentation rate, raised C- reactive protein (CRP), and other acute inflammatory markers (23). In cases of severe disease, blood cultures may be positive. Ultrasonography of the thyroid may reveal an abscess. The latter requires aspiration and pus should be sent for microbiological diagnosis. Typical findings of acute suppurative thyroiditis on ultrasound are perithyroidal hypoechoic space, effacement of the plane between the thyroid and surrounding tissues, and unilateral presentation [Fig 1] (27). Computed Tomography (CT) offers better spatial resolution and can be used in cases where ultrasound is not showing characteristic findings or when there is involvement of nearby soft tissue structures. Barium swallow studies may be required to diagnose a pyriform sinus, especially in children when there are recurrent episodes of suppurative thyroiditis (28).

Fig 1. A. Ultrasound of the thyroid showing enlargement of the left lobe of the thyroid with heterogenous echotexture, suggestive of thyroiditis B. Ultrasound Doppler showing increased vascularity of the left lobe of the thyroid

Aspiration or surgical drainage of pus with intravenous empirical broad-spectrum antibiotics (especially in sick patients) is the cornerstone of management for acute suppurative thyroiditis. If the patient is immunocompromised, antifungal therapy should be added to initial therapy. In case of extensive involvement of nearby structures, surgical debridement of involved areas may be needed. With respect to culture sensitivity, antibiotic therapy can be modified and once clinical improvement occurs, patients can be switched to oral antibiotics. If there is presence of pyriform fistula, it should be treated either surgically (removal of entire tract with thyroidectomy) or by ablation (21, 29).

 

Subacute Thyroiditis

 

Subacute thyroiditis (also termed as granulomatous, giant cell, or deQuervain’s thyroiditis), is usually due to a viral illness following respiratory illness. Rarely, bacterial infections like Mycobacterium tuberculosis, Treponema pallidum, or Yersinia enterocolitica may cause subacute thyroiditis. Tuberculous thyroiditis is discussed in another chapter (2). Differentiating features of subacute thyroiditis and suppurative thyroiditis are presented in table 3 (19, 30, 31).

 

Table 3. Differentiating Acute Suppurative Thyroiditis and Subacute Thyroiditis

Features

Acute suppurative thyroiditis

Sub-acute thyroiditis

Etiology

Usually bacterial in origin

Usually follows viral upper respiratory tract infection

Presentation

Rapidly evolving, patient can be very toxic with extensive involvement

Presents with systemic symptoms over days to week

Age

Children, 20 to 40 years

20 to 60 years

Sex

Slight female preponderance

More common in females

Fever

 72%

54%

Neck pain

 70%

77%

Neck tenderness

Usually, unilateral (Left sided involvement due to persistent pyriform sinus)

Bilateral and migratory

Redness over skin

Common

Not present

Swelling with fluctuation suggestive of abscess formation

 Common

Not present

History of sore throat

Absent

Present

Clinical features of thyrotoxicosis

Not common

Common in the initial phase

Laboratory

 

 

Leukocytosis

82%

25 to 50 %

Raised ESR

90%

85%

Abnormal thyroid function test

44%

60%

FNAC

Pus

Giant cells, granulomas

Ultrasound Thyroid

Hypoechoic areas with abscess formation, usually unilateral

Ill-defined hypoechoic areas, usually in bilateral lobes

RAIU study

Normal

Decreased in the initial thyrotoxic phase

18 F FDG PET

Increased uptake

Increased uptake

CT scan

Useful when ultrasound is doubtful and when infection extends into peri thyroid tissue

Not useful

Treatment

Antibiotics & drainage of pus

NSAIDS, glucocorticoids in severe cases and sequential follow up of thyroid function tests.

FNAC- fine needle aspiration cytology; RAIU- radioactive iodine uptake; NSAIDS-Non steroidal anti-inflammatory drugs

 

BACTERIAL INFECTIONS OF ADRENALS

 

Tuberculosis of Adrenals

 

Tuberculosis of the adrenal glands is the most common cause of primary adrenal insufficiency in developing countries. An autoimmune etiology remains common in developed countries. Tuberculous infection of the adrenal gland occurs from hematogenous spread from pulmonary or genitourinary sites (32). Adrenals are the most common endocrine gland involved in tuberculosis (2). The symptoms are usually non-specific with generalized weakness, easy fatiguability, loss of weight, loss of appetite, pain in abdomen, and gradually progressive darkening of complexion (Fig 2). These symptoms and signs of adrenal insufficiency do not occur until more than 90% of the glands are destroyed (33). Patient can have low grade fever if the tuberculosis is active and cough and hemoptysis if associated pulmonary involvement is present. In the majority of the cases, the tuberculosis infection may not be active with only a past history of pulmonary tuberculosis (33). Untreated patients may present with adrenal crisis during times of stress. Laboratory investigations reveal low serum cortisol and high plasma adrenocorticotrophic hormone (ACTH). Sometimes, ACTH stimulation test (short synacthen test) may be needed. Adrenal insufficiency is ruled out if serum cortisol level one hour post synacthen (ACTH) stimulation is more than 500-550 nmol/L (14-20 ug/dL depending on the assay). Electrolyte abnormalities noted in adrenal insufficiency are hyponatremia and hyperkalemia. Computed tomography shows bilateral enlarged adrenal masses with areas of necrosis and caseation. In long standing cases, there may be evidence of calcifications (33). Diagnosis is confirmed by adrenal biopsy showing caseating granulomas with acid fast bacilli. Other methods like culture and molecular techniques can be used for diagnosing tuberculosis in biopsy samples. Anti-tubercular treatment (ATT) along with both glucocorticoid and mineralocorticoids remain the treatment of choice. ATT consists of isoniazid - INH (5 mg/kg /d), rifampicin (10 mg/kg /d), pyrazinamide (30 mg/kg /d), and ethambutol (20 mg/kg/d) for 3 to 6 months, subsequently isoniazid and rifampicin for 6 to 12 months (34). In case of multi drug resistant tuberculosis, ATT may be altered with respect to the pattern of resistance. It may require second line medications and longer duration of therapy. Patients usually require lifelong replacement therapy with glucocorticoids and mineralocorticoids.

 

Apart from Mycobacterium tuberculosis, in the context of HIV-AIDS and other immunocompromised states, Mycobacterium avium intracellular and Mycobacterium chelonae may also cause primary adrenal insufficiency.

Fig 2. A. Darkening of the skin in the dorsum aspect of hands in a patient with primary adrenal insufficiency due to adrenal tuberculosis B. Darkening of the palmar aspect including palmar creases of the same patient

Adrenal Abscess

 

An adrenal abscess is a rare clinical condition with very few cases reported in the literature. Organisms that are implicated are Mycobacterium, anaerobes, Salmonella, Nocardia, and E coli. Treatment includes drainage of abscess and antibiotic therapy (35-40). The choice of antibiotic is guided by culture and sensitivity report. In culture negative cases, broad spectrum antibiotics with coverage for gram positive, gram negative, and anaerobic organisms should be considered.

 

Waterhouse-Friderichsen Syndrome

 

Waterhouse-Friderichsen syndrome (WFS) or purpura fulminans is an uncommon clinical entity associated with bilateral adrenal hemorrhage in the setting of severe bacterial sepsis, which was first reported by Rupert Waterhouse and Carl Friderichsen (41). The initial version of this syndrome was classically described with Neisseria meningitidis sepsis. But later it was found that a similar clinical picture was seen with other bacterial infections such as Streptococcus pneumoniaeHemophilus influenzae, Escherichia coli, Staphylococcus aureus, Group A beta-hemolytic Streptococcus, Capnocytophaga canimorsus, Enterobacter cloacae, Pasteurella multocida, Plesiomonas shigelloides,Neisseria gonorrhoeaeMoraxella duplex, Rickettsia rickettsia, Bacillus anthracis, Treponema pallidum,and Legionella pneumophila (42-44).

 

Adrenal glands are predisposed to hemorrhage because around 50-60 small adrenal branches from 3 main adrenal arteries form a subcapsular plexus that drains into the medullary sinusoids through only a few venules (43). Therefore, an increase in adrenal venous pressure due to any cause may lead to hemorrhage. These bacteria may invade the adrenals directly or may produce endotoxins to cause adrenal necrosis and hemorrhage. There is also evidence of microthrombi within the adrenals along with disseminated intravascular coagulation (DIC). Pathologically, organisms are hardly demonstrated in the adrenal specimens (45). The patients are usually sick and present with profound adrenal crisis and shock.  A petechial rash is usually present on the trunk, lower limbs, and mucous membrane and its severity correlates with the degree of thrombocytopenia (44). Treatment involves admission to an intensive care unit and resuscitation with intravenous fluids, intravenous glucocorticoids, and appropriate antibiotics.

 

BACTERIAL INFECTIONS OF GONADS

 

Bacterial Infections of Testes

 

EPIDEMIOLOGY

 

Infection of the epididymis can occur in both children and adults. In severe cases, the inflammation can spread further into testis and present as epididymo-orchitis. If the duration of illness is less than 6 weeks, it is termed as acute epididymo-orchitis, whereas duration more than 6 weeks is termed as chronic. In children, it usually occurs between two and thirteen years of age, whereas in adults, it is common between twenty and thirty years of age (46).

 

BACTERIOLOGY

 

Causative organisms in younger males less than 35 years of age are Neisseria gonorrhoeae and Chlamydia trachomatis. In older men, causative organisms include Escherichia coli, other coliforms, and Pseudomonas. Rare bacterial causes include Ureaplasma species, Mycoplasma genitalium, Mycobacterium tuberculosis, and Brucella species (47-49). Risk factors for epididymitis include urinary tract infections, sexually transmitted diseases, bladder outlet obstruction, prostate enlargement, and urinary tract surgeries or urogenital procedures. In homosexual men, an enteric bacterial etiology is common (46, 50).

 

CLINICAL PRESENTATION

 

Acute epididymitis presents as localized testicular pain. On palpation, there may be swelling in the posterior part of the testis that represents an enlarged testis and inflamed epididymis. More advanced cases present with secondary testicular pain and swelling (epididymo-orchitis). There could be redness of scrotum and hydrocele (reactive fluid collection secondary to infection) (Fig 3). A positive Prehn sign (manual elevation of the scrotum relieves pain) is more often seen with epididymitis than testicular torsion (46).

 

Fig 3. Swelling of bilateral testes with reddening of the skin overlying the scrotum, suggestive of epidymo-orchitis

 

DIAGNOSIS AND MANAGEMENT

 

In all cases of acute epididymo-orchits, it is important to rule out acute surgical conditions like testicular torsion and Fournier’s gangrene. All patients should undergo routine urine microscopy, urine for culture and sensitivity, and a urine nucleic acid amplification test (NAAT) for N. gonorrhoeae and C. trachomatis. NAAT is helpful in diagnosing infections where urine cultures are negative (51). Management depends on the severity of illness, history suggestive of sexually transmitted diseases, and reports of NAAT (summarized in table 4) (46, 52).

 

Table 4. Management of Acute Epididymo-Orchitis

Clinical scenario

Likely organisms

Choice of empirical antibiotic therapy *

Children < 14 years

Various possibilities – secondary to anatomical issues

Treatment based on urine culture results and referral to urologist.

Individuals at risk of sexually transmitted diseases but do not practice anal intercourse

N. gonorrhoeae and C. trachomatis 

 

Single injection of ceftriaxone 500mg intramuscular and oral Doxycycline 100mg twice daily for 10 days. 

 

Alternative for doxycycline – Azithromycin

Alternative for ceftriaxone- Gentamycin

 

Individuals at risk of sexually transmitted diseases but do practice anal intercourse

N. gonorrhoeae, C. trachomatis and enteric pathogens

 

Single injection of ceftriaxone 500mg intramuscular and oral Doxycycline 100mg twice daily for 10 days plus oral levofloxacin 500 mg once daily for 10 days

 

 

Individuals at lower risk of sexually transmitted diseases

Recent urinary tract surgery or instrumentation

 

Enteric pathogens

Oral levofloxacin 500 mg once daily for 10 days

 

*Further treatment should be adjusted based culture and NAAT results; severe cases may require hospitalization and intravenous antibiotics.

 

Bacterial Infections of Ovaries

 

Isolated infection of ovaries is not common. It is usually part of pelvic inflammatory disease. In severe cases, it may present as tubo-ovarian abscess. Tubo-ovarian abscesses are often polymicrobial and typically contain a predominance of anaerobic bacteria. Common organisms include Escherichia coli, Bacteroides fragilis, other Bacteroides species, Pepto-streptococci, and anaerobic streptococci (53). Diagnosis is based on history, physical examination, ultrasound suggesting tubo -ovarian mass or abscess, and microbiological diagnosis. Treatment consists of admission, intravenous antibiotic therapy, and aspiration of abscess if needed. Patients who do not respond, will need surgical intervention (54).

 

 

Yersinia enterocolitica has been implicated in the pathogenesis of autoimmune thyroid disease (55). Immunoglobulins from patients with Yersinia infection inhibit binding of TSH to thyrocytes (56). This could be explained by structural similarity between Yersinia outer membrane proteins (YOP) and epitopes of the TSH receptor (55, 56).

 

Role of gut microbiome has recently implicated in the metabolic syndrome, obesity, and diabetes (57). Many metabolites produced by gut microbes get absorbed into the circulation. They may act on specific receptors to regulate metabolism (58, 59). Also, some bacterial components can act as endocrine factors controlling metabolism(58).

 

CONCLUSION

 

Bacterial infections of the endocrine glands are rare. Pituitary abscesses usually occur in the setting of underlying pathology of the pituitary gland.  It is commonly caused by Streptococci and Staphylococci. MRI of the sella demonstrates a characteristic rim of enhancement after gadolinium injection. Treatment of pituitary abscess is trans-sphenoidal surgery and intravenous antibiotics. Culture is positive in only 19.7% of cases. Acute suppurative thyroiditis is commonly caused by Streptococci and Staphylococci. Important risk factor for acute suppurative thyroiditis in children is pyriform fistula, whereas in adults, it is more common in immunocompromised states. Acute suppurative thyroiditis appear as hypoechoic area on ultrasound. It is treated by ultrasound guided drainage of the abscess and antibiotic therapy. Acute suppurative thyroiditis should be differentiated from subacute thyroiditis. Primary adrenal bacterial infections other than tuberculosis are rare. Waterhouse-Friderichsen syndrome (WFS) is an uncommon clinical entity associated with bilateral adrenal hemorrhage in the setting of severe bacterial sepsis. It is classically described with Neisseria meningitides, but may be associated with other bacteria as well. Toxins produced by bacteria can cause necrosis, hemorrhage, and microthrombi within the adrenal gland leading to WFS. Infection of the epididymis can occur in both children and adults. Sometimes, the inflammation spreads further into testis and presents as epididymo-orchitis. Common bacterial agents causing epididymo-orchitis are N. gonorrhoeae and C.trachomatis. Enteric pathogens should be suspected if there is history of homosexual practice. Management depends on the severity of illness, history of suggestive sexually transmitted diseases, and reports of NAAT (urine nucleic acid amplification test).

 

ACKNOWLEDGEMENTS

 

Dr. Lovekesh Bhatia, Department of Radiodiagnosis, Aadhar Health Institute, Hisar, India

Dr. Vinita Jain, Department of Pediatrics, Aadhar Health Institute, Hisar, India

 

REFERENCES

 

  1. Nagendra L, Boro H, Mannar V. Bacterial Infections in Diabetes. 2022 Apr 5. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, et al. editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000.
  2. Gupta S, Ansari MAM, Gupta AK, Chaudhary P, Bansal LK. Current Approach for Diagnosis and Treatment of Adrenal Tuberculosis-Our Experience and Review of Literature. Surg J (N Y). 2022 Mar 3;8(1):e92-e97.
  3. Jacob JJ, Paul PAM. Infections in Endocrinology: Tuberculosis. 2021 Mar 14. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, et al. editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000.
  4. Machado MJ, Ramos R, Pereira H, Barbosa MM, Antunes C, Marques O, Almeida R. Primary pituitary abscess: case report and suggested management algorithm. Br J Neurosurg. 2021 Aug 24:1-4.
  5. Pekic S, Miljic D, Popovic V. Infections of the Hypothalamic-Pituitary Region. 2021 Aug 9. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, et al. editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000.
  6. Cabuk B, Caklılı M, Anık I, Ceylan S, Celik O, Ustün C. Primary pituitary abscess case series and a review of the literature. Neuro Endocrinol Lett. 2019 Oct;40(2):99-104.
  7. Pekic S, Popovic V. Alternative causes of hypopituitarism: traumatic brain injury, cranial irradiation, and infections. Handb Clin Neurol. 2014;124:271-90.
  8. Adams D, Kern PA. A case of pituitary abscess presenting without a source of infection or prior pituitary pathology. Endocrinol, Diabetes & Metabol Case Reports [Internet]. 2016 Aug 16 [cited 2022 May 10];2016.
  9. Gao L, Guo X, Tian R, Wang Q, Feng M, Bao X, Deng K, Yao Y, Lian W, Wang R, Xing B. Pituitary abscess: clinical manifestations, diagnosis and treatment of 66 cases from a large pituitary center over 23 years. Pituitary. 2017 Apr;20(2):189-194.
  10. Ling X, Zhu T, Luo Z, Zhang Y, Chen Y, Zhao P, et al. A review of pituitary abscess: our experience with surgical resection and nursing care. Transl Cancer Res. 2017 Aug;6(4):852–9.
  11. Vates GE, Berger MS, Wilson CB. Diagnosis and management of pituitary abscess: a review of twenty-four cases. J Neurosurg. 2001 Aug;95(2):233-41.
  12. Bloomer ZW, Knee TS, Rubin ZS, Hoang TD. Case of an atypical pituitary abscess. BMJ Case Rep. 2021 Nov 30;14(11):e246776. 
  13. Bricaire L, Van Haecke C, Laurent-Roussel S, Jrad G, Bertherat J, Bernier M, Gaillard S, Groussin L, Dupin N. The Great Imitator in Endocrinology: A Painful Hypophysitis Mimicking a Pituitary Tumor. J Clin Endocrinol Metab. 2015 Aug;100(8):2837-40.
  14. Benzick AE, Wirthwein DP, Weinberg A, Wendel GD Jr, Alsaadi R, Leos NK, Zeray F, Sánchez PJ. Pituitary gland gumma in congenital syphilis after failed maternal treatment: a case report. Pediatrics. 1999 Jul;104(1):e4. 
  15. Spinner CD, Noe S, Schwerdtfeger C, Todorova A, Gaa J, Schmid RM, Busch DH, Neuenhahn M. Acute hypophysitis and hypopituitarism in early syphilitic meningitis in a HIV-infected patient: a case report. BMC Infect Dis. 2013 Oct 17;13:481.
  16. Har-el G, Sasaki CT, Prager D, Krespi YP. Acute suppurative thyroiditis and the branchial apparatus. Am J Otolaryngol. 1991 Jan-Feb;12(1):6-11. 
  17. Touihmi S, Rkainilham, Mehdaoui A, El boussaadni Y, Oulmaati A. Acute suppurative thyroiditis with abscess. J Pediatr Surg Case Rep. 2021 Feb;65:101757.
  18. Bukvic B, Diklic A, Zivaljevic V. Acute suppurative klebsiella thyroiditis: a case report. Acta Chir Belg. 2009 Mar-Apr;109(2):253-5.
  19. Paes JE, Burman KD, Cohen J, Franklyn J, McHenry CR, Shoham S, Kloos RT. Acute bacterial suppurative thyroiditis: a clinical review and expert opinion. Thyroid. 2010 Mar;20(3):247-55.
  20. Madana J, Yolmo D, Kalaiarasi R, Gopalakrishnan S, Saxena SK, Krishnapriya S. Recurrent neck infection with branchial arch fistula in children. Int J Pediatr Otorhinolaryngol. 2011 Sep;75(9):1181-5. 
  21. Parida PK, Gopalakrishnan S, Saxena SK. Pediatric recurrent acute suppurative thyroiditis of third branchial arch origin--our experience in 17 cases. Int J Pediatr Otorhinolaryngol. 2014 Nov;78(11):1953-7.
  22. Zhang P, Tian X. Recurrent neck lesions secondary to pyriform sinus fistula. Eur Arch Otorhinolaryngol. 2016 Mar;273(3):735-9
  23. Lafontaine N, Learoyd D, Farrel S, Wong R. Suppurative thyroiditis: Systematic review and clinical guidance. Clin Endocrinol (Oxf). 2021 Aug;95(2):253-264
  24. Falhammar H, Wallin G, Calissendorff J. Acute suppurative thyroiditis with thyroid abscess in adults: clinical presentation, treatment and outcomes. BMC Endocr Disord. 2019 Dec 3;19(1):130.
  25. Dunham B, Nicol TL, Ishii M, Basaria S. Suppurative thyroiditis. Lancet. 2006 Nov 11;368(9548):1742.
  26. Minhas SS, Watkinson JC, Franklyn J. Fourth branchial arch fistula and suppurative thyroiditis: a life-threatening infection. J Laryngol Otol. 2001 Dec;115(12):1029-31.
  27. Masuoka H, Miyauchi A, Tomoda C, Inoue H, Takamura Y, Ito Y, Kobayashi K, Miya A. Imaging studies in sixty patients with acute suppurative thyroiditis. Thyroid. 2011 Oct;21(10):1075-80.
  28. Furukawa M, Kano M, Takiguchi T, Umeda R. Piriform sinus fistula as a route of infection in acute suppurative thyroiditis. Auris Nasus Larynx. 1986;13(2):107-12.
  29. Miyauchi A, Inoue H, Tomoda C, Amino N. Evaluation of chemocauterization treatment for obliteration of pyriform sinus fistula as a route of infection causing acute suppurative thyroiditis. Thyroid. 2009 Jul;19(7):789-93. 
  30. Pearce EN, Farwell AP, Braverman LE. Thyroiditis. N Engl J Med. 2003 Jun 26;348(26):2646-55.
  31. Shrestha RT, Hennessey J. Acute and Subacute, and Riedel’s Thyroiditis. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, et al., editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000-.
  32. Del Borgo C, Urigo C, Marocco R, Belvisi V, Pisani L, Citton R, et al. Diagnostic and therapeutic approach in a rare case of primary bilateral adrenal tuberculosis. J Med Microbiol. 2010 Dec;59 (Pt 12):1527-1529
  33. Kelestimur F. The endocrinology of adrenal tuberculosis: the effects of tuberculosis on the hypothalamo-pituitary-adrenal axis and adrenocortical function. J Endocrinol Invest. 2004 Apr;27(4):380-6.
  34. Laway BA, Khan I, Shah BA, Choh NA, Bhat MA, Shah ZA. Pattern of adrenal morphology and function in pulmonary tuberculosis: response to treatment with antitubercular therapy. Clin Endocrinol (Oxf). 2013 Sep;79(3):321-5.
  35. Regino CA, Gómez JP, Mosquera-Klinger G. Endoscopic Ultrasound-Guided Transgastric Puncture and Drainage of an Adrenal Abscess in an Immunosuppressed Patient. Clin Endosc. 2022 Mar;55(2):302-304. 
  36. O'NEILL JA Jr, HALL WH. ISOLATED ADRENAL ABSCESS SECONDARY TO SALMONELLA. Arch Surg. 1965 Mar;90:454-6.
  37. Midiri M, Finazzo M, Bartolotta TV, Maria MD. Nocardial adrenal abscess: CT and MR findings. Eur Radiol. 1998;8(3):466-8.
  38. Yokoyama S, Sekioka A, Utsunomiya H, Hara S, Takahashi T, Yoshida A. Adrenal abscess as a complication of Escherichia coli sepsis in neonates: A case report. J Pediatr Surg Case Rep. 2013 Sep;1(9):328–30.
  39. Jin W, Miao Q, Wang M, Zhang Y, Ma Y, Huang Y, et al. A rare case of adrenal gland abscess due to anaerobes detected by metagenomic next-generation sequencing. Ann Transl Med. 2020 Mar;8(5):247. 
  40. Rumińska M, Witkowska-Sędek E, Warchoł S, Dudek-Warchoł T, Brzewski M, Pyrżak B. Adrenal abscess in a 3-week-old neonate - a case report. J Ultrason. 2015 Dec;15(63):429-37.
  41. Varon J, Chen K, Sternbach GL. Rupert Waterhouse and Carl Friderichsen: adrenal apoplexy. J Emerg Med. 1998 Jul-Aug;16(4):643-7. 
  42. Hamilton D, Harris MD, Foweraker J, Gresham GA. Waterhouse-Friderichsen syndrome as a result of non-meningococcal infection. J Clin Pathol. 2004 Feb;57(2):208-9. 
  43. Karki BR, Sedhai YR, Bokhari SRA. Waterhouse-Friderichsen Syndrome. [Updated 2021 Dec 12]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2022 Jan-.
  44. Kalinoski T. Waterhouse-Friderichsen Syndrome with Bilateral Adrenal Hemorrhage Associated with Methicillin-Resistant Staphylococcus aureus (MRSA) Bacteremia in an Adult Patient with History of Intravenous Drug Use. Am J Case Rep. 2022 Apr 14;23:e936096.
  45. Guarner J, Paddock CD, Bartlett J, Zaki SR. Adrenal gland hemorrhage in patients with fatal bacterial infections. Mod Pathol. 2008 Sep;21(9):1113-20.
  46. McConaghy JR, Panchal B. Epididymitis: An Overview. Am Fam Physician. 2016 Nov 1;94(9):723-726. 
  47. Tracy CR, Steers WD, Costabile R. Diagnosis and management of epididymitis. Urol Clin North Am. 2008 Feb;35(1):101-8; vii. 
  48. Doble A, Taylor-Robinson D, Thomas BJ, Jalil N, Harris JR, Witherow RO. Acute epididymitis: a microbiological and ultrasonographic study. Br J Urol. 1989 Jan;63(1):90-4. 
  49. Hawkins DA, Taylor-Robinson D, Thomas BJ, Harris JR. Microbiological survey of acute epididymitis. Genitourin Med. 1986 Oct;62(5):342-4.
  50. Kaver I, Matzkin H, Braf ZF. Epididymo-orchitis: a retrospective study of 121 patients. J Fam Pract. 1990 May;30(5):548-52.
  51. Wampler SM, Llanes M. Common scrotal and testicular problems. Prim Care. 2010 Sep;37(3):613-26, x.
  52. Trojian TH, Lishnak TS, Heiman D. Epididymitis and orchitis: an overview. Am Fam Physician. 2009 Apr 1;79(7):583-7.
  53. Kairys N, Roepke C. Tubo-Ovarian Abscess. 2021 Jul 18. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2022 Jan–.
  54. Munro K, Gharaibeh A, Nagabushanam S, Martin C. Diagnosis and management of tubo-ovarian abscesses. Obstet Gynecol. 2018 Jan;20(1):11–9.

         Mar;93(3):674-6.

  1. Effraimidis G, Wiersinga WM. Mechanisms in endocrinology: autoimmune thyroid disease: old and new players. Eur J Endocrinol. 2014 Jun;170(6):R241-52. 
  2. Li R, Li Y, Li C, Zheng D, Chen P. Gut Microbiota and Endocrine Disorder. Adv Exp Med Biol. 2020;1238:143-164.
  3. Rastelli M, Cani PD, Knauf C. The Gut Microbiome Influences Host Endocrine Functions. Endocr Rev. 2019 Oct 1;40(5):1271-1284
  4. Cani PD, Knauf C. How gut microbes talk to organs: The role of endocrine and nervous routes. Mol Metab. 2016 May 27;5(9):743-52.

Hyperglycemic Hyperosmolar State

CLINICAL RECOGNITION

 

The hyperglycemic hyperosmolar state (HHS) is a life-threatening metabolic decompensation of diabetes which presents with severe hyperglycemia and profound dehydration, typically accompanied by alteration in consciousness ranging from lethargy to coma. In contrast to diabetic ketoacidosis (DKA) in which acidemia and ketonemia are key features, these are limited in HHS. Mortality in HHS ranges from 5-20% and is higher at the extremes of age and in the presence of coma. HHS is more prevalent in type 2 diabetics and in about 7-17% of cases is the initial presentation classically seen in institutionalized elderly patients with diminished thirst perception or inability to ambulate to get free water as needed. HSS is extremely rare as first presentation in patients with type 1 diabetes. Infections are the leading precipitant of HHS, but it can also be precipitated by poor medication compliance, cerebrovascular accident, myocardial infarction, pancreatitis, trauma, alcohol abuse and drugs such as corticosteroids and atypical antipsychotics.

 

PATHOPHYSIOLOGY

 

HHS and DKA represent the two ends of the spectrum of markedly decompensated diabetes, differing mainly in severity of acidosis, ketosis and dehydration. HHS usually occurs with a lesser degree of insulinopenia compared with DKA, but the pathophysiology is otherwise thought to be similar. In both entities, there is a decrease in net effective insulin action with concomitant elevation of counterregulatory hormones. In the setting of relative insulin deficiency, glucagon, catecholamines and cortisol stimulate hepatic glucose production though glycogenolysis and gluconeogenesis. High catecholamines and low insulin reduce peripheral glucose uptake. Unlike DKA, there is adequate insulin available in HHS to restrain lipolysis and ketogenesis, as well as to restrain marked elevation of counterregulatory hormones, such cortisol, glucagon and growth hormone. However, there is significant hyperglycemia with resultant glycosuria leading to loss of water and electrolytes, dehydration, decreased renal perfusion, decreased glucose clearance, and exacerbation of hyperglycemia, ultimately causing impaired level of consciousness. In HHS, the initial increase in proinflammatory cytokines, reactive oxygen species, and plasminogen activator inhibitor-1 can contribute to increased prothrombotic risk.

 

DIAGNOSIS AND DIFFERENTIAL

 

HHS usually has a slower onset than DKA, with symptoms developing over several days or weeks. Patients present with polyuria, polydipsia, weakness, and blurred vision. Altered sensorium is classic in HHS, but mental status can range from fully alert to confused, lethargic, or comatose. Seizure can occur in up to 20% of the patients. Exam reveals physical signs of dehydration, including dry mucous membranes, poor skin turgor, cool extremities, hypotension, and tachycardia. Fever may or may not be present, suggesting underlying infection, although normothermia or even hypothermia may be present due to concomitant vasodilatation.

 

The diagnostic criteria for HHS include severe hyperglycemia and hyperosmolality with preservation of near normal pH and bicarbonate, and minimal or absent serum and/or urine ketones. ADA guidelines note glucose level at presentation should be > 600 mg/dl, with pH > 7.3 and bicarbonate level > 20 mEq/L. These are common diagnostic criteria that differentiate HHS from DKA (Table 1). However, it is clear that a subpopulation of patients with type 2 diabetes can present with overlapping features of HHS and DKA. Patients with ketosis prone type 2 diabetes present with ketosis and milder acidosis than the one expects in DKA and in some cases with near normal pH and bicarbonate. More rarely, HHS can present in the setting of diabetes insipidus where patients are treated with intravenous dextrose for the severe dehydration leading to hyperglycemia and glycosuria.

 

Table 1. Diagnosis of HHS Versus DKA

 

HHS

DKA

Diagnostic criteria

pH

>7.30

≤7.30

Plasma Glucose

>600 mg/dl

>250 mg/dl

Serum bicarbonate

>15 mEq/L

<18 mEq/L

Plasma and urine ketones

None or trace

Positive

Anion gap

 

<12

>12

Serum Osmolality

>320 mOsm/kg

Variable

Glycosuria

 

++

++

Typical Deficit

Water (ml/kg)

100-200 (9L)

100 (6L)

Na+ (mEq/kg)

5-13

7-10

Cl- (mEq/kg)

5-15

3-5

K+(mEq/Kg)

4-6

3-5

P04 (mmol/kg)

3-7

5-7

Mg++& Ca++(mEq/kg)

1-2

1-2

Adapted from Kitabchi A, et al. Diabetes Care, 2006, 29: 2739-2747

 

DIAGNOSTIC TESTS NEEDED

 

Initial laboratory tests should include electrolytes with calculated anion gap, plasma glucose, blood urea nitrogen (BUN), creatinine, serum and urine ketones, osmolality, and arterial blood gas.  Evidence of infection should be sought by checking complete blood count with differential and urinalysis, with consideration of additional evaluation including chest X-ray, and culture of blood, sputum and urine. Electrocardiogram and head CT should be done if clinically indicated.  HHS produces significant loss of several electrolytes as well as a prerenal azotemia and increased hematocrit, the latter due to hemoconcentration. An increase of serum sodium in the presence of hyperglycemia indicates severe dehydration. Altered mentation appears to correlate with the degree of hyperosmolality; hence significantly diminished mentation in the setting of an osmolality of <320 mOsm/kg should prompt a search for other causes. It is notable that despite significant potassium losses, serum potassium is usually normal or even elevated on presentation because of extracellular shift in the setting of hyperosmolality and insulin deficiency. HgbA1c may be useful to discriminate chronic uncontrolled hyperglycemia from acute metabolic decompensation.   

 

THERAPY

 

There is a lack on randomized controlled trials for the treatment of HHS and the American Diabetes Association (ADA) has developed guidelines that combine the treatment of HHS and DKA. The treatment of HHS includes a four-pronged approach:

  • reestablishment of volume status with vigorous intravenous hydration;
  • electrolyte replacement;
  • correction of hyperglycemia with volume expansion and administration of intravenous insulin;
  • diagnosis and management of potential precipitants.

 

The initial emergent treatment has been summarized in table 2.

 

Fluid Replacement

 

Aggressive fluid replacement is critical in order to prevent cardiovascular collapse, with repletion of intravascular and extravascular volume and restoration of renal perfusion. The total fluid deficit should be estimated (usually 100-200 ml/kg), with the goal of replacement over 24 hours. In the absence of heart failure, 1-1.5 liters of isotonic saline should be given over the first hour. Subsequent fluid replacement depends on the hydration and electrolyte status. In patients with hypotension, aggressive isotonic saline infusion should continue until the patient is stabilized. Increased plasma sodium concentration in the setting of hyperglycemia suggests a significant water deficit; clinical practice guidelines recommend adding a correction factor of 1.6 mg/dl to the plasma sodium concentration for each 100 mg/dl of glucose above 100 mg/dl. In the normotensive patient with a corrected serum sodium level that is normal or high, fluid replacement can be continued with half normal saline given at 250-500 cc/hour, whereas if the corrected serum sodium level is low, isotonic saline should be administered at similar rate. When serum glucose reaches 200-300 mg/dl, fluid should be changed to 5% dextrose solution in half normal saline.

 

Electrolyte Replacement

 

Electrolyte replacement is the second crucial step in HHS management. Serum potassium can be normal or elevated on presentation despite total body potassium depletion. Osmotic-induced intracellular dehydration results in potassium efflux from the cells. Since insulin causes a shift of potassium into the cell, it is mandatory to correct the potassium level to >3.3 mEq/L before starting insulin therapy. If potassium is between 3.3 and 5.3 mEq/L, 20-30 mEq of potassium should be given in each liter of intravenous fluid to keep serum potassium between 4 to 5 mEq/L. The potassium should be monitored if >5.3 meq/L and potassium replacement initiated when potassium < 5.3 meq/L. Magnesium should be checked and repleted as necessary; this is important to prevent renal wasting of potassium with exacerbation of hypokalemia. Routine administration of phosphate is not recommended (17); however, careful phosphate replacement can be considered in patients with very low levels (<1 meq/L), cardiac dysfunction, or respiratory distress.

 

Insulin Therapy

 

The treatment of choice for correction of hyperglycemia is regular insulin by continuous infusion after adequate fluid and potassium replacement. While randomized controlled studies in patients with DKA have shown that insulin therapy is effective regardless of the route of administration, there is limited data supporting the use of subcutaneous or intramuscular insulin in HHS and continuous intravenous insulin administration remains the treatment of choice in patient with significant dehydration, reduced level of consciousness, and critical illness. Insulin should be given as initial bolus of 0.1 unit per kilogram body weight, followed by a drip of 0.1 unit per kilogram per hour; alternatively, 0.14 units per kilogram per hour can be given as an infusion without a bolus. If the glucose level does not decrease by 50-70 mg/dl in the first hour, the insulin dose may be doubled.  When the plasma glucose level reaches 300 mg/dl, insulin infusion may be reduced to 0.05-0.1 unit/kg/hour and dextrose can be added to the fluids to keep the glucose level between 250-300 mg/dl until hyperosmolality has resolved and the patient is alert.

 

Evaluation of Precipitant Factors

 

Evaluation for and treatment of potential precipitant factors is important. Patients with HHS have a mortality rate of about 5-20%, 10-fold higher than patients with DKA and several studies have shown that the increased mortality is likely because of the precipitating factors. For this reason, appropriate work up and treatment should be given as indicated.

 

Table 2. Initial Emergent Treatment for HHS

1--IV Fluids

a-Cardiogenic shock

b-Severe hypovolemia

c-Mild dehydration

Hemodynamic Monitoring/ Pressors/ 0.9% NaCl

0.9% NaCl (1L/hr.)

Na* low:

0.9% NaCl (250-500 ml/hr.) † 
Na* normal or high:

0.45% NaCl (250-500 ml/hr.) † 

 

When serum glucose ≤300 mg/dl, 5% dextrose/0.45% NaCl (150-250 ml/hr.)

2-IV Potassium (with adequate renal function)

 

a--K+ <3.3 mEq/L

b--K+ 3.3-5.3 mEq/L

c--K+ >5.3 mEq/L

Hold insulin, K 20-30 mEq/ hr. until K+ >3.3 mEq/L

K 20-30 mEq in each liter of IV fluid to keep K+ 4-5 mEq/L

Do not give K; monitor

3-IV Insulin

 

Bolus 0.1 unit/Kg, then 0.1 unit/Kg/hr. infusion (or 0.14 unit/kg/hr. infusion w/o bolus)

Double infusion if glucose does not decrease by 50-70 mg/dl in the first hour

When serum glucose 300 mg/dl, ↓ insulin infusion to 0.05-0.1 units/Kg/hr.

Adapted from Kitabchi A, et al. Diabetes Care, 2006, 29: 2739-2747

*Corrected serum sodium; † depending on the hydration status

 

FOLLOW-UP

 

Meticulous clinical and laboratory follow up is critical in patients with HHS. Capillary blood glucose levels should be monitored every hour to allow adjustment of the insulin infusion. Electrolytes, BUN, creatinine and plasma glucose should be checked every 2-4 hours until the patient is stable. When plasma osmolality is <315 mOsm/L, and the patient is alert and able to eat, a multidose insulin regime consisting of long-acting insulin and short/rapid acting insulin before meals may be initiated. Intravenous insulin infusion should be continued for 1-2 h after the subcutaneous insulin is given to ensure adequate plasma insulin levels are maintained.

 

It is also important to monitor for possible treatment-related complications, the most common of which are hypoglycemia and hypokalemia.  These are both usually due to overzealous treatment with insulin and can be minimized with frequent monitoring. Cerebral edema is extremely rare in patients with HHS, and usually occurs in younger adults. To reduce the risk of cerebral edema in high-risk patients, sodium, glucose, and water deficit may be more gradually corrected to avoid the rapid decline in plasma osmolality. Recurrence of HHS can be prevented by improved patient as well as caregiver education and enhanced access to medical care. For elderly nursing home residents, nursing home staff should be educated in recognition of signs and symptoms of HHS and on the importance of adequate fluid intake.

 

REFERENCES

 

            Gosmanov AR, Gosmanova EO, Kitabchi AE. Hyperglycemic Crises: Diabetic Ketoacidosis and Hyperglycemic Hyperosmolar State. 2021 May 9. 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, Laferrère 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, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–. PMID: 25905280

 

Pasquel, F.J., and Umpierrez, G.E. 2014. Hyperosmolar hyperglycemic state: a historic review of the clinical presentation, diagnosis, and treatment. Diabetes Care 37:3124-3131.

 

 

Umpierrez, G., and Korytkowski, M. 2016. Diabetic emergencies - ketoacidosis, hyperglycaemic hyperosmolar state and hypoglycaemia. Nat Rev Endocrinol 12:222-232.

 

Kitabchi, A.E., Umpierrez, G.E., Miles, J.M., and Fisher, J.N. 2009. Hyperglycemic crises in adult patients with diabetes. Diabetes Care 32:1335-1343.

 

Palmer, B.F., and Clegg, D.J. 2015. Electrolyte and Acid-Base Disturbances in Diabetes Mellitus. N Engl J Med 373:2482-2483.

 

Dhatariya, K.K., and Vellanki, P. 2017. Treatment of Diabetic Ketoacidosis (DKA)/Hyperglycemic Hyperosmolar State (HHS): Novel Advances in the Management of Hyperglycemic Crises (UK Versus USA). Curr Diab Rep 17:33.

 

 

 

 

 

 

 

 

 

Initial Management of Severe Hyperglycemia in Type 2 Diabetes

CLINICAL RECOGNITION

 

Type 2 diabetes mellitus (DM) is a common disease affecting 26 million people, 8.3% of the US population.  Of these, an estimated 7 million people are undiagnosed.

 

Type 2 DM typically has two pathophysiologic defects:  an insulin secretory defect and insulin resistance.  Symptoms of uncontrolled hyperglycemia include polyuria, polydipsia, blurry vision, and possibly dehydration and weight loss. Patients may complain of thirst, sweet cravings, generalized fatigue, abdominal discomfort, and muscle cramps. They may have a history of poor wound healing and/or frequent infections. Basic metabolic laboratory tests may reveal a random blood glucose level over 200 mg/dL [11.1 mmol/L], hyper- or hyponatremia, hypokalemia, metabolic acid-base derangements, and acute renal or prerenal insufficiency. Historical clues for the diagnosis of type 2 DM might include pre-existing history of pre-diabetes, a family history of type 2 diabetes, an ethnicity at higher risk for DM (African-American, Hispanic, Native American, Pacific Islander), a history of gestational diabetes, obesity, and sedentary lifestyle. 

 

PATHOPHYSIOLOGY  

 

Table 1. Clinical Features of the Acute Presentation of Type 2 Diabetes and Pathophysiology

Hyperglycemia

Insulin resistance, insulin deficiency (pancreatic beta cell failure), increased gluconeogenesis, glycogenolysis

Dehydration, polyuria, polydipsia

Osmotic diuresis, compensatory thirst

Weight loss, sweet cravings

Glycosuric calorie loss and inadequate glucose utilization

Muscle pain and abdominal discomfort

Lactic acid accumulation, hypokalemia, electrolyte /acid-base derangements

Metabolic alkalosis and/or acidosis, electrolyte disturbances

Dehydration and ketogenesis

Ketogenesis

Insulin deficiency resulting in lipolysis yielding free fatty acids, substrate for formation of ketone bodies

 

DIAGNOSIS AND DIFFERENTIAL

 

Diabetes can be diagnosed in several ways: 1) Presence of symptoms of hyperglycemia with a random blood glucose of 200 mg/dL [11.1 mmol/L]; 2) fasting blood glucose > 126 mg/dL [7.0 mmol/L; 3) the 75-gram oral glucose tolerance test with a blood glucose > 200 mg/dL [11.1 mmol/L] at 2 hours; 4) hemoglobin A1C value > 6.5%.  If asymptomatic, the diagnosis of diabetes is confirmed with two consecutive day abnormal results from the same test or a different test or with two different tests on the same day. If using the hemoglobin A1C for diagnosis, one should be aware of several conditions (some common) making this measure un-interpretable

 

Adult patients with type 1 and type 2 DM can sometimes present similarly.  If a patient presents with hyperglycemia, ketonemia, and metabolic acidosis, distinguishing between types of diabetes is not necessary in this acute setting because initially, both type 1 and type 2 DM are treated with insulin.  Later the two diseases may be distinguished with antibody testing although this is neither completely sensitive nor specific. Type 2 DM can also present acutely with a hyperglycemic hyperosmolar state (HHS) with dehydration, altered level of consciousness, and a lesser degree of clinical ketosis than seen in diabetic ketoacidosis (DKA). Consideration for genetic syndromes and concomitant rare conditions of endocrine hormone excess (cortisol, growth hormone, epinephrine, glucagon) leading to hyperglycemia should be in the non-urgent setting for patients with new diagnoses of diabetes.

 

DIAGNOSTIC TESTS NEEDED AND SUGGESTED

 

For an acute presentation of diabetes with hyperglycemic symptoms, the patient should have a basic metabolic panel of laboratory tests including glucose, electrolytes, blood urea nitrogen, creatinine, blood and or urinary ketones, liver function tests, and urinalysis. Other testing should be guided by a patient’s history and physical exam and might include evaluation for infection or cardiac dysfunction.  A hemoglobin A1C reflects the average blood glucose over the last 90 days and is a helpful test.  Distinguishing type 1 from type 2 DM can on occasions be difficult but can be assisted with autoantibody testing [tyrosine phosphatase antibody (IA-2) or glutamic acid decarboxylase (GAD) 65 antibody]. The presence of antibody suggests an autoimmune lesion as seen in type 1 DM. In type 1 DM insulin and C-peptide levels are characteristically low, whereas they may be normal or elevated at the onset of type 2 DM.

 

TREATMENT

 

Insulin therapy is the initial management choice for patients presenting with hyperglycemia and catabolic symptoms including weight loss. If laboratory abnormalities suggest concurrent DKA or HHS, these must be treated emergently with aggressive saline rehydration, intravenous insulin, potassium and other electrolyte replacement.

 

For a severely hyperglycemic patient, with a catabolic presentation that usually includes moderate to severe volume depletion, the first therapeutic step is rehydration, usually with intravenous saline.  After adequate hydration, therapy with physiologic doses of insulin (0.3-0.4 units per kilogram body weight daily) is recommended. The ideal treatment regimen would be a combination of a long-acting basal insulin plus multiple premeal prandial “bolus” injections to manage meal-related insulin requirements and correction of pre-meal hyperglycemia, referred to as basal-bolus insulin therapy. A good starting place is to prescribe half the total daily insulin dose as basal and the other half as bolus. The combination of long-acting insulin and a rapid acting analogue are good options for basal-bolus therapy. The basal dose is given as a separate injection from the bolus injection.

 

The premeal “bolus” dose is calculated by summing the dose required to cover the carbohydrate load plus the dose to correct premeal hyperglycemia and is given as one injection 10-15 minutes before the meal. Particularly with premeal hyperglycemia but even with mealtime glucose levels within target, today’s rapid-acting analogues require time for absorption to avoid more severe postprandial hyperglycemia (this is typically called the “lag time”).  In an acute setting, and in a less sophisticated patient, it might be more appropriate to begin therapy with a twice-daily pre-mixed insulin. Even though this regimen is not ideal for many for the long-term because it does not allow for sufficient dose titration, this regimen allows approximate physiologic basal-bolus insulin coverage with fewer injections. Nevertheless, if starting with basal-bolus or premixed insulin, it is best to teach the patient to use the strategy of correcting pre-meal hyperglycemia with an additional dose of rapid acting insulin analogue, given 10-15 minutes before the meal. This adds tremendous flexibility to an otherwise rigid regimen.

 

Until more education is possible, the need to limit high glycemic-load carbohydrate intake (such as with sweetened beverages and juice) should be strongly reinforced with counseling. Certainly, arrangements for general and dietary diabetes education should be made for a newly diagnosed diabetic patient or for a patient new to insulin therapy.

 

FOLLOW-UP

 

The patient will use a glucose meter to check his/her fasting and premeal blood glucose levels.  For the patient on basal-bolus insulin therapy, he/she will increase bedtime basal insulin doses by 1-2 units every 3 days until fasting blood glucose falls into target range of 90 -130 mg/d [5 – 7.2 mmol/L]. Ideally, bedtime and fasting glucose levels are about the same at the end of the basal insulin titration. If there is a consistent reduction in bedtime to fasting glucose by more than 50 mg/dL [2.8 mmol/L], basal insulin dose is too high.

 

Adjustments for pre-meal insulin doses are most easily made with an algorithm written clearly for the patient to reference. The importance of injecting the mealtime insulin 10 -15 minutes before eating needs to be emphasized. In contrast to type 1 diabetes where carbohydrate counting is standard, most type 2 patients do well by taking the same mealtime dose or altering up or down based on the size of the meal. For example, one might take 8 units for a smaller meal and 12 units for a large one. If patients feel hypoglycemic symptoms (sweating, shaking, mental fogginess, hunger) despite concurrent blood glucoses levels in the normal range, one could use smaller insulin dose increments to lower blood glucose into the target range more gradually.  Generally, increases of insulin dose by10% are well tolerated by patients.  Late night snacks without insulin coverage may lead to morning hyperglycemia and interfere with the assessment of the adequacy of the bedtime insulin doses.  Correction doses are “trial and error” but most patients with type 2 diabetes require an “insulin sensitivity factor” of 30 (i.e., 30 mg/dL glucose reduction expected from one unit of insulin injected). For example, if additional insulin is provided for premeal glucose levels above 150 mg/dL, 1 extra unit would be given for 150-180 mg/dL, 2 units for 181-210 mg/dL, etc.  When starting insulin, it may be appropriate to use a more conservative insulin sensitivity factor such as 40 or 50.

 

Table 2.  Premeal Bolus Dose Calculation Using Rapid-Acting Insulin Analogue

Total premeal insulin dose is sum of:

Suggested Units

Meal coverage

5-8 units for smaller meal, 9-12 units for larger meal

Pre meal hyperglycemia correction

1 unit per 30-50 mg/dL above150 mg/dL

 

Initial diabetes therapy includes counseling for lifestyle and diabetic nutritional interventions.  Starting therapy with metformin could also be considered as an adjunctive therapy with insulin to reduce insulin requirements and minimize weight gain. Overtime with lifestyle changes, a decrease in glucose toxicity, and the addition of other hypoglycemic agents some patients who present with very high glucose levels may be able discontinue insulin therapy.

 

GUIDELINES

 

2019 Update to: Management of Hyperglycemia in Type 2 Diabetes, 2018. A Consensus Report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Buse JB, Wexler DJ, Tsapas A, Rossing P, Mingrone G, Mathieu C, D'Alessio DA, Davies MJ. Diabetes Care. 2020 Feb;43(2):487-493

 

REFERENCES

 

Donner T, Sarkar S. Insulin – Pharmacology, Therapeutic Regimens, and Principles of Intensive Insulin Therapy. 2019 Feb 23. 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, Laferrère 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, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–. PMID: 25905175

 

Feingold KR. Oral and Injectable (Non-Insulin) Pharmacological Agents for the Treatment of Type 2 Diabetes. 2021 Aug 28. 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, Laferrère 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, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–. PMID: 25905364

 

Gosmanov AR, Gosmanova EO, Kitabchi AE. Hyperglycemic Crises: Diabetic Ketoacidosis and Hyperglycemic Hyperosmolar State. 2021 May 9. 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, Laferrère 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, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–. PMID: 25905280

 

 

Diabetic Ketoacidosis

CLINICAL RECOGNITION

 

Omission of insulin and infection are the two most common precipitants of diabetic ketoacidosis (DKA). Noncompliance may account for up to 44% of DKA presentations; while infection is less frequently observed.

 

Acute medical illnesses involving the cardiovascular system (myocardial infarction, stroke, acute thrombosis), gastrointestinal tract (bleeding, pancreatitis), endocrine axis (acromegaly, Cushing`s syndrome, hyperthyroidism) and recent surgical procedures can contribute to the development of DKA by causing dehydration, increase in insulin counterregulatory hormones, and worsening of peripheral insulin resistance.

 

Medications such as diuretics, beta-blockers, corticosteroids, second-generation anti-psychotics, anti-convulsants, sodium-glucose cotransporter-2 (SGLT-2) inhibitors, and/or immune checkpoint inhibitors may affect carbohydrate metabolism and volume status and, therefore, could precipitate DKA. SGLT-2 inhibitors have been associated with euglycemic DKA (glucose level < 250mg/dL)

 

Other factors leading to DKA include psychological problems, eating disorders, insulin pump malfunction, and drug abuse. It is well recognized that new onset T2DM can sometimes manifest with DKA. These patients are obese, mostly African Americans or Hispanics and have undiagnosed hyperglycemia, impaired insulin secretion, and impaired insulin action. A recent report suggests that cocaine abuse is an independent risk factor associated with DKA recurrence.

 

PATHOPHYSIOLOGY

 

Insulin deficiency, increased insulin counter-regulatory hormones (cortisol, glucagon, growth hormone, and catecholamines), and peripheral insulin resistance lead to hyperglycemia, dehydration, ketosis, and electrolyte imbalance which underlie the pathophysiology of DKA.

 

Hyperglycemia of DKA evolves through accelerated gluconeogenesis, glycogenolysis, and decreased glucose utilization – all due to absolute insulin deficiency. Of note, diabetes patients who developed DKA while treated with SGLT-2 inhibitors can present without hyperglycemia, i.e., with euglycemic DKA.

 

Due to increased lipolysis and decreased lipogenesis, abundant free fatty acids are converted to ketone bodies: β-hydroxybutyrate (β-OHB), acetoacetate, and acetone. Hyperglycemia-induced osmotic diuresis, if not accompanied by sufficient oral fluid intake, leads to dehydration, hyperosmolarity, electrolyte loss, and subsequent decrease in glomerular filtration. With decline in renal function, glycosuria diminishes and hyperglycemia/hyperosmolality worsens. With impaired insulin action and hyperosmolality, utilization of potassium by skeletal muscle is markedly diminished leading to intracellular potassium depletion. Also, potassium is lost via osmotic diuresis causing profound total body potassium deficiency. Therefore, DKA patients can present with broad range of serum potassium concentrations. Nevertheless, a “normal” plasma potassium concentration may indicate that potassium stores in the body are severely diminished and the institution of insulin therapy and correction of hyperglycemia will lead to hypokalemia.

 

DIAGNOSIS AND DIFFERENTIAL DIAGNOSIS

 

Diagnostic criteria for DKA are presented in Table 1.

 

Table 1. Criteria and Classification of DKA

DKA

Mild

Moderate

Severe

Plasma glucose (mg/dl)

>250 mg/dl

>250mg/dl

>250mg/dl

Arterial pH

7.25-7.30

7.00-7.24

<7.00

Serum bicarbonate (mEq/L)

15-18

10- 15

<10

Urine ketone*

+

+

+

Serum ketone*

+

+

+

Effective Serum Osmolality**

Variable

Variable

Variable

Anion Gap***

>10

>12

>12

Mental Status

Alert

Alert/drowsy

Stupor/coma

*Nitroprusside reaction method

** Serum osmolality: 2[measured Na+ (mEq/L)] + glucose (mg/dl)/18 = mOsm/kg

*** Anion Gap: [ (Na+)– (Cl- + HCO3- (mEq/L)]

 

CLINICAL PRESENTATION

 

Polyuria, polydipsia, weight loss, vomiting, and abdominal pain usually are present in patients with DKA. Abdominal pain can be closely associated with acidosis and resolves with treatment. Physical examination findings such as hypotension, tachycardia, poor skin turgor, and weakness support the clinical diagnosis of dehydration in DKA. Mental status changes may occur in DKA and are likely related to degree of acidosis and/or hyperosmolarity. A search for symptoms of precipitating causes such as infection, vascular events, or existing drug abuse should be initiated in the emergency room. Patients with hyperglycemic crises can be hypothermic because of peripheral vasodilation and decreased utilization of metabolic substrates.

 

DIFFERENTIAL DIAGNOSIS

 

Hyperglycemic hyperosmolar state is not associated with ketosis. Starvation and alcoholic ketoacidosis are not characterized by hyperglycemia >200 mg/dl and bicarbonate level <18 meq/L. With hypotension, decreased renal function, and history of metformin use, lactic acidosis (lactic acid level >7 mmol/L) should be suspected. Ingestion of methanol, isopropyl alcohol, and paraldehyde can also alter anion gap and/or osmolality and need to be investigated.

 

Table 2. Laboratory Evaluation of Causes of Acidosis

Factor Studied

DKA

HHS

Starvation

Uremic acidosis

pH

normal

normal

Mild↓

Plasma glucose

>500 mg/dl

normal

normal

Glycosuria

+ +

+ +

0

0

Total plasma ketones*

↑↑

0 or ↑

Mild↑

0

Anion gap

Normal

Mild↑

Mild↑

Osmolality

>330 mOsm/kg

normal

Normal/↑

Other

     

BUN>200 mg/dl

HHS- hyperglycemic hyperosmolar state

BUN –blood urea nitrogen

*Acetest and Ketostix (Bayer; Leverkusen, Germany) measure acetoacetic acid only; thus, misleadingly low values may be obtained because the majority of “ketone bodies” are β-hydroxybutyrate.

 

DIAGNOSTIC TESTS NEEDED

 

Initial Necessary Tests

 

Basic metabolic panel, osmolality, ketones, β-hydroxybutyrate (β-OH), complete blood count with differential, urinalysis and urine ketones by dipstick, and arterial blood gases.

 

Additional Tests

 

Electrocardiogram, chest X-ray, and various tissue cultures, if indicated, and HbA1c.

 

Caveats to Diagnostic Tests

 

Anion gap acidosis is calculated by subtracting the sum of Cl and HCO3 from measured (not corrected) Na concentration and should be corrected for hypoalbuminemia. Usually, a HCO3 level of 18-20 meq/L rules out metabolic acidosis. Arterial blood gases with pH<7.30 support the diagnosis. β-OHB is early and abundant ketoacid and indicative of ketosis. Acetoacetate but not acetone, is a product of ketone body formation and is measured by a nitroprusside reaction that is widely used but may be negative in the blood in early DKA. Effective serum osmolality can be measured directly or derived from following formula: 2 x [measured Na+(meq/L)] + glucose/18. High measured Na indicates a significant degree of dehydration. A white blood cell count >25,000 should warrant a comprehensive search for infection. Serum creatinine can be falsely elevated because of acetoacetate interference with the colorimetric creatinine assay. When patients with DKA present with mixed acid-base disorder, measurement of serum β-OHB will be required to confirm that acidosis is due to ketoacidosis.

 

THERAPY 

 

The therapeutic goals of management include optimization of:

  • volume status,
  • hyperglycemia and ketosis/acidosis,
  • electrolyte abnormalities,
  • potential precipitating factors.

 

Steps to follow in early stages of DKA management (Figures 1, 2, 3):

  • Start IV fluids after blood sample for biochemistry was sent to laboratory (Fig. 1);
  • Potassium level should be >3.3 meq/L before initiation of insulin therapy (supplement potassium intravenously if needed) (Fig. 3);
  • Initiate insulin therapy only when steps 1-2 are executed (Fig. 2).

 

Resolution of DKA:

  • Plasma glucose <200-250 mg/dl,
  • Serum bicarbonate concentration >18 meq/L,
  • Venous blood pH >7.3, and
  • Anion gap <10

 

Fluid therapy: Replace fluid deficit in DKA (~6 L) within 24-36 hours with the goal of 50% volume replacement within first 12 hours.

 

Insulin Therapy: Transition to subcutaneous insulin by giving long-acting insulin 2 hours before the discontinuation of IV insulin.

 

Bicarbonate therapy: If pH is < 7.0 or bicarbonate level is < 5 meq/L, administer 100 mmol (2 ampules) of bicarbonate in 200 ml of water with 20 meq of potassium chloride over two hours.

Figure 1. Fluid management in adult patients with DKA

 

Figure 2. Insulin management in adult patients with DKA

Figure 3. Potassium management in adult patients with DKA

 

FOLLOW UP: COMPLICATIONS AND DISCHARGE

 

Hypoglycemia and hypokalemia are the most frequent complications and can be prevented by timely adjustment of insulin dose and frequent monitoring of potassium levels.

 

Non-anion gap hyperchloremic acidosis occurs due to urinary loss of ketoanions which are needed for bicarbonate regeneration and preferential re-absorption of chloride in proximal renal tubule secondary to intensive administration of chloride-containing fluids and low plasma bicarbonate. The acidosis usually resolves and should not affect treatment course.

 

Cerebral edema is reported in young adult patients. This condition is manifested by appearance of headache, lethargy, papillary changes, or seizures. Mortality is up to 70%. Mannitol infusion and mechanical ventilation should be used to treat this condition.

 

Rhabdomyolysis is another possible complication due to hyperosmolality and hypoperfusion.

 

Pulmonary edema can develop from excessive fluid replacement in patients with CKD or CHF.

 

Discharge planning should include diabetes education, selection of appropriate insulin regimen that is understood and afforded by the patient, and preparation of set of supplies for the initial insulin administration at home.

 

REFERENCES

 

Kitabchi AE, Umpierrez GE, Fisher JN, Murphy MB, and Stentz FB. Thirty years of personal experience in hyperglycemic crises: diabetic ketoacidosis and hyperglycemic hyperosmolar state. The Journal of clinical endocrinology and metabolism 93: 1541-1552, 2008.http://www.ncbi.nlm.nih.gov/pubmed/18270259

 

Kitabchi AE, Umpierrez GE, Miles JM, and Fisher JN. Hyperglycemic crises in adult patients with diabetes. Diabetes care 32: 1335-1343, 2009.

 

Karslioglu French E, Donihi AC, Korytkowski MT. Diabetic ketoacidosis and hyperosmolar hyperglycemic syndrome: review of acute decompensated diabetes in adult patients.

BMJ. 2019 May 29;365:l1114.

 

Gosmanov AR, Gosmanova EO, Kitabchi AE. Hyperglycemic Crises: Diabetic Ketoacidosis and Hyperglycemic Hyperosmolar State. 2021 May 9. 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, Laferrère 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, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–. PMID: 25905280

Diabetes Management During Ramadan

ABSTRACT

Muslims contribute to 25% of the world population and majority of them reside in the diabetes and obesity endemic Asia-Pacific region. Fasting during Ramadan is one of the five pillars of Islam and an obligatory duty for all healthy adolescents and adult Muslims. However, Islam exempts the ill and pregnant women from fasting. Despite this, many individuals with diabetes who are at high risk from fasting, fast during Ramadan. Individuals fasting during Ramadan are less likely to see their physicians before starting the fast and more likely to fast against medical advice. Hence, these individuals are at increased risk of hyperglycemia, hypoglycemia, and cardiovascular and renal complications. Management of diabetes during Ramadan needs a comprehensive and integrated planning and dissemination of knowledge through the healthcare providers and Muslim religious leaders. Diabetes care should start in the pre-Ramadan period, continue through Ramadan, and follow-up in the post-Ramadan period.

 

INTRODUCTION

 

Muslims contribute to approximately 25% of world population and are distributed across >200 countries across the globe (1,2). Of this, 61.7% of Muslims live in Asia-Pacific region, which is also a region experiencing the diabetes epidemic (3,4).

 

Ramadan is the 9th holy month of the Islamic lunar calendar. Fasting during Ramadan is one of the five pillars of Islam (5). Fasting during Ramadan is an obligatory duty for all healthy adolescents and adult Muslims aimed at spiritual and holistic wellbeing of the individual (1,5). The Holy Quran exempts the sick, medically unfit, or those traveling from fasting during the holy month (1,5).

 

 

Fasting during Ramadan involves complete abstinence from food, medication, drink (including water), or any other form of nutrition (including via a percutaneous endoscopic gastrostomy tube) from dawn to sunset (1, 2, 6).  The fasting during Ramadan is a type of intermittent fasting as it is observed for 10–21 hours depending on the geographical location and solar season and is observed daily for 29–30 consecutive days (1, 2). Individuals fasting during Ramadan take two main meals, Suhoor (pre-dawn meal) and Iftar (post sunset meal) and eat nothing from sunrise to sunset (1, 2, 6).

 

It is estimated that about 79% of Muslims with type 2 diabetes (T2D) and about 43% of them with type 1 diabetes (T1D) fast during Ramadan (7). Of those who fast during Ramadan, 64% fasted every day, and 94.2% fasted for at least 15 days (8). The medication timings of these individuals with diabetes need to be adjusted to pre-dawn and post-sunset timings (1,2). Also, many of these individuals fast against medical advice (9, 10).

 

Since a huge proportion of individuals with diabetes fast during Ramadan, and many are at risk due to fasting, management of diabetes during Ramadan and proper fasting guidance is critical (1,2). 

 

EFFECTS OF FASTING DURING RAMADAM

 

Physiological Changes

 

Fasting during Ramadan is associated with a number of physiological changes.

 

CHANGES IN FEEDING PATTERNS AND ENERGY INTAKE

 

Ramadan fasting differs from other forms of fasting as there is no consumption of any food or drink between dawn and sunset. Hence, the timing between the meals is very long, and this disrupts the normal physiology with disruption in the normal rhythm and fluctuations seen in various homeostasis and endocrine processes (Figure 1). Major changes occur in glucose homeostasis in individuals with diabetes that results in post Iftar hyperglycemia and risk of hypoglycemia during the day (Figures 2 and 3)

Figure 1. Changes in feeding patterns and energy intake during various fasting periods (11, 12). (I) normal feeding, (II) Ramadan fasting and (III) prolonged fasting and starvation.

 Figure 2. Mean continuous glucose monitoring (CGM) profiles from healthy individuals (12, 13).

Figure 3. Mean continuous glucose monitoring (CGM) profiles from individuals with diabetes fasting during Ramadan (12, 13).

DECREASE IN TOTAL SLEEP TIME

Total sleep time decreases by approximately 1 hour, with a decrease in sleep period time, rapid eye movement (REM) sleep proportion and duration. Additionally, delayed sleep and an increase in non-REM sleep proportion, sleep latency, and daytime sleepiness by1-point on the Epworth sleepiness scale is also observed (ESS) (12).

ALTERATION OF CIRCADIAN RHYTHM AND HORMONE LEVELS

Sudden alteration of circadian rhythm and hormone levels occurs due to sudden changes in sleep and wake cycles and feeding patterns. Fasting can induce epigenetic changes in genes that control the circadian rhythm (12). The change in circadian rhythm triggers many catametabolic changes, alteration in temperature, and changes in the normal rhythm of hormones like insulin, glucagon, leptin, ghrelin, cortisol, melatonin, growth hormone, and testosterone (12,14). The various changes seen are:

 

  • Insulin resistance and increased glucagon levels: excessive glycogen breakdown and increased gluconeogenesis
  • Cortisol circadian rhythm shows a shift with a blunting of the morning to evening ratio. However, serum cortisol levels do not change by end of Ramadan month.
  • Morning adiponectin levels are reduced
  • Morning and evening growth hormone levels are reduced
  • Large increases in morning leptin levels
  • No major shifts in diurnal ghrelin level

 

By the end of Ramadan significant decrease in serum levels of ghrelin, leptin, and melatonin are observed along with modest reductions in testosterone in men.

SHIFT IN FLUID BALANCE

A sudden shift in fluid balance is seen because of an absolute restriction of fluid intake between dawn and sunset. This may precipitate dehydration in a hot climate which may in turn cause hypotension and falls (6). Uncontrolled hyperglycemia can exacerbate the dehydration due to an osmotic diuresis (6). Dehydration in individuals with T2D can present as low blood pressure, lethargy, or syncope. Dehydration can also increase the risk of thrombosis and stroke due to hemoconcentration and hypercoagulability (6). Other fluid related changes are not considered a major cause of concern and include higher fluid and total water intake between sunset and dawn; urine osmolality increases significantly in the afternoon to conserve water and reduce urine output (12).

ALTERED ENERGY BALANCE

Altered energy balance is seen due to a sudden increase in food intake at Iftar. During Ramadan there is a reduction in activity and energy expenditure which is offset by the reduced time spent during sleep (12).

GUT MICROBIOTA

Intermittent fasting during Ramadan can have direct impact on the gut microbiota which could lead to positive changes in health (12).

 LIPID CHANGES

Fasting during Ramadan has been shown to be associated with a significant increase in high-density lipoprotein-cholesterol (HDL-C) and a significant decrease in total triglycerides, total cholesterol, and low-density lipoprotein-cholesterol (LDL-C) (6, 15).

Physical and Mental Wellbeing

Fasting during Ramadan can have both positive and negative effects on the physical and mental wellbeing of the individuals (Table 1) (16).

Table 1. Positive and Negative Effects on Physical and Mental Wellbeing of Individuals Fasting During Ramadan (16)

Positive benefits

Negative effects

Sense of fulfilment

Sleep deprivation and disruption of circadian rhythm leading to an increase in fatigue and reduction in cognition

More lethargy 

Improvements in:

Weight and BMI

Self-control and ability to resist temptations

Glucose excursions causing feelings of being unwell

Greater sense of:

Empathy for less fortunate

Community

Fostering relationships

Heightened feelings of fear for diabetes related complications

Participation in Sunnah practices for greater spiritual benefits

Temporary changes in weight

Reducing potentially harmful vices, such as smoking, for greater physical and mental wellbeing

Short term feelings of stress anxiety, irritability, and agitation

BMI- body mass index

 

The month long fasting during Ramadan has been associated with significant reduction in weight, waist circumference, and fat mass, especially in those who are overweight or obese (15, 17).

 RISKS OF FASTING DURING RAMADAN IN INDIVIDUALS WITH DIABETES

The various risks of fasting in individuals with diabetes who fast during Ramadan are:

  • Hyperglycemia
  • Hypoglycemia
  • Macrovascular: Cardiovascular disease (CVD) including stroke
  • Microvascular: Chronic kidney disease (CKD)
  • Dehydration

Dual Risk of Hyperglycemia and Hypoglycemia

In people with diabetes fasting during Ramadan there can be an increase in glucose variability and therefore there is increased risk of both hyperglycemia and hypoglycemia (12).

HYPERGLYCEMIA

The meals at Iftar are calorie dense and can cause a significant and rapid rise in blood glucose (BG) levels in people with diabetes (12). The EPIDIAR study showed that the hospitalization rate for severe hyperglycemia during Ramadan increased significantly in individuals with T2D (P<0.001). The hospitalization rate for severe hyperglycemia (with or without ketoacidosis) during Ramadan increased insignificantly for individuals with T1D (P = 0.1635) (6,7).

HYPOGLYCEMIA

The CREED study showed that hypoglycemia incidence before Ramadan was associated with significantly increased risk of hypoglycemia during Ramadan (18). This association between hypoglycemia incidence before and during Ramadan has been seen through multiple studies across continents (1,18,19). Similarly, the EPIDAR study7 showed that T1D and T2D patients had a 4.7-fold and a 7.5-fold increase, respectively, in severe hypoglycemia requiring hospitalization during Ramadan. Hypoglycemia during Ramadan was significantly associated with the use of sulfonylureas and insulin (18,19). Severe and non-severe hypoglycemia rates are fewer with second-generation sulfonylureas, Glucagon-like peptide-1 receptor agonists (GLP-1 RAs), insulin analogues, and sodium-glucose transporter-2 (SGLT-2) inhibitors (20-25). During the early Ramadan period, patients on sulfonylureas and those on ≥2 antidiabetic medications have significant increase in mean amplitude of glycemic excursions (26).

 

Other factors influencing the incidence of hypoglycemia during Ramadan include season, geographical location, fasting duration, time since diagnosis, gender, anthropometric measures, dietary behaviors, and pre-fasting education (1). 

Macrovascular (Cardiovascular and Cerebrovascular) and Microvascular Risk (Renal Complications)

Diabetes increases the risk of CVD and stroke, and individuals with diabetes who have pre-existing CVD or stroke are at greater risk of complications when fasting (27). Individuals with unstable CVD or stroke are also at very high risk from fasting. Individuals with diabetes and CKD stage 3 are at high-risk from fasting while those with stage 4-5 are at very high risk from fasting during Ramadan (27). Patients on dialysis or those who had a kidney transplant are also considered high risk from fasting (27).

 

Factors associated with increased risk of fasting during Ramadan are high carbohydrate intake, inadequate hydration, high activity levels, poor sleeping patterns, and missing doses of essential medicines (27).

 

All these high to very high-risk individuals should be discouraged from fasting. If they still decide to fast, then pre-Ramadan assessment and education and monitoring during Ramadan and post-Ramadan should be carried out under the expert guidance of a multidisciplinary team (diabetologist, cardiologist, neurologist, nephrologist, nutritionist etc.) (27). Weekly monitoring during Ramadan by a health care provider should be encouraged.

 

Pre-Ramadan assessment and education should begin three months prior to Ramadan and all efforts should be made to stabilize the doses of the various drugs, adjust them to morning and evening dose, and those on insulin should be taught self-titration of dose based on SMBG (27).

Patients with CKD, on dialysis or those who had a kidney transplant should be encouraged to routinely monitor electrolytes and creatinine at various time-points during Ramadan (27). A diet rich in potassium and phosphorus should be avoided (27).

 MANAGEMENT OF DIABETES DURING RAMADAN

5 R's of Ramadan Care

 

The management of diabetes during Ramadan and in general can be summarized under the mnemonic termed as the 5 R's of Ramadan care (Table 2).

 

Table 2. The 5 R's of Ramadan Care (28)

The 5 R

Significance

Respect

Respect the patient's attitudes, wishes, and needs, and consider these while planning therapy

Speak with the patient with empathy for his religious beliefs

Risk stratification

This is an essential backbone for pre-Ramadan counseling

Revision of Therapy

Diabetes therapy will need to be revised based on risk of hyperglycemia and hypoglycemia, and other risk factors

Regular Follow Up

Regular follow-up with HCP before, during and after Ramadan is necessary to ensure a safe and healthy fasting experience

Reappraisal of Strategy

Diabetes is a dynamic condition, and constant reappraisal is required during the current and next fasting period

HCP- health care provider

Pre-Ramadan Management of Diabetes

DM management in people planning to fast during Ramadan should ideally start six to eight or maximum 12 weeks before the first day of fasting (6, 29). Diabetes assessment and plan of care pre-Ramadan should ideally follow the flow chart shown in Figure 4 (6, 29).

Figure 4. Diabetes assessment and plan of care pre-, during and post Ramadan (6, 29)

 

Patient education during this time-period is necessary because many patients follow a self-management approach of diabetes during Ramadan and do not appreciate the risks and implications of fasting on DM and its medications, and fast against medical advice (6, 19). Physicians need to be sensitized about this time-period for their Muslim patients as many may not realize the religious sensitivities associated with DM management during Ramadan (6).

PRE-RAMADAN EDUCATION

The pre-Ramadan diabetes education should cover:

Risk quantification, exemptions, and removing misconceptions

  • Blood glucose monitoring
  • Fluids and dietary advice
  • Physical activity and exercise advice
  • Medication adjustment
  • When to break the fast
  • Recognition of hypoglycemia and hyperglycemia symptoms

RISK STRATIFICATION OF PEOPLE WITH DIABETES

The pre-Ramadan time period should be used to understand the individual’s risks associated with fasting, and develop an individualized treatment plan for the individual who falls in the lower risk category and can fast. The risk stratification is done based on several factors (6, 9, 10):

  • Type and duration of diabetes
  • DM treatment regime and polypharmacy with multiple glucose lowering drugs
  • Level of glycemic control
  • Risk or occurrence of hypoglycemia
  • DM self-management capability including hypoglycemia awareness, motivation for self-monitoring blood glucose (SMBG), frailty, and cognition
  • DM complications and comorbidities
  • Ongoing/recent severe illness
  • Renal impairment
  • Social determinants affecting assess and adherence to treatment including economic and education level
  • For those with T1D: access to continuous glucose monitoring (CGM) and advanced insulin technologies

 

The IDF-DAR Practical Guidelines 2021 stratify the individuals with DM who are going to fast during Ramadan into three risk categories, low, moderate and high (Figure 5) (29).

Figure 5. IDF-DAR 2021 Ramadan risk score and risk categories for fasting (2, 30)

 

DM patients who had a history of severe hypoglycemia ≤3 months before Ramadan, recurrent hypoglycemia, or of hypoglycemia unawareness are considered high risk (31). Young individuals and adolescents start fasting during Ramadan with the onset of puberty, and those with T1D are considered to be high risk for fasting and generally discouraged from fasting (9).

 

Many DM patients may need to be upgraded to high-risk category during the current coronavirus-2019 (COVID-19) pandemic and thus likely to fall under the ‘advised not to fast’ risk category (6).

 

Patients with high-risk scores are advised not to fast as fasting is not considered safe for these individuals (29). However, since fasting during Ramadan is a personal choice, the individuals deciding to fast despite being cautioned against it, should be monitored very closely during and after Ramadan (6, 19).

 

Individuals with low and moderate risk scores are educated about the risks and advised to fast with strict BG monitoring, and adjustments to diet/nutrition and medications (29). The individuals are closely monitored during Ramadan by their health care providers (HCPs) and advised to come for a post Ramadan follow-up. At this follow-up visit, the HCPs asses the glycemic control, and discuss the challenges faced to make the next Ramadan fasting more risk free. Pre-Ramadan risk assessment, education and advise is known to improve the fasting experience of individuals with DM (6).

Blood Glucose Monitoring

 

Self-monitoring blood glucose (SMBG) should be stressed upon and encouraged. Moderate to low-risk individuals with diabetes can monitor their BG once or twice daily but those at high risk of fasting should be encouraged to follow a 7-time-point guide for SMBG during Ramadan (Figure 6) (29). Additional BG check should be done if the individual experiences symptoms of hypoglycemia, hyperglycemia, or feel unwell (29).

 

Figure 6. A seven time-point blood glucose monitoring guide for people with diabetes fasting during Ramadan (29)

Fluids and Dietary Advice

Detailed diet plan for Ramadan is provided through Ramadan specific medical nutrition therapy (MNT) and Ramadan Nutrition Plan (RNP) (29,32). Adequate fluid intake should be encouraged between sunset and dawn.

Physical Activity and Exercise Advice

Individuals with diabetes who are fasting should be encouraged to carry out their normal physical activity (29). Taraweeh prayers, which involve activities such as bowing, kneeling, and rising, can be considered as part of daily exercise activities (29). Rigorous exercise/activity during the fasting period should be avoided as this can increase the risk of hypoglycemia and dehydration, especially during the last few hours of fasting (29).

Medication Adjustment

All medications (diabetes and non-diabetes) should be reviewed in the pre-Ramadan period to see which medications need a dose and time adjustment. The change should be made well before Ramadan and monitored through appropriate clinical and laboratory evaluation (29). Patients on insulin should be taught self-titration of units based on SMBG values (10).

When to Break the Fast

This is an essential component of Pre-Ramadan education. All individuals with diabetes who are fasting during Ramadan should be advised to break their fast if:

  • Blood glucose <70 mg/dL (3.9 mmol/L)
  • Advise to re-check within 1 hour if BG is between 70–90 mg/dL (3.9–5.0 mmol/L)
  • Blood glucose >300 mg/dL (16.6mmol/L)
  • Symptoms of hypoglycemia, hyperglycemia, dehydration or acute illness occur

Recognizing Symptoms of Hypoglycemia and Hyperglycemia

All individuals with diabetes who are fasting during Ramadan and their caregivers should be taught to recognize symptoms of hypoglycemia and hyperglycemia (Figure 7) (29). If they recognize these symptoms, they should be advised to break their fast.

Figure 7. Symptoms of hypoglycemia and hyperglycemia (29)

 The Medico-Religious Interplay in Ramadan

Muslims believe that Ramadan is a blessed month, and see fasting during the holy month of Ramadan as a deeply meaningful and spiritual experience (1, 30). A significant number of individuals with diabetes fast during Ramadan, even against medical advice and despite the religious exemptions available to the sick (1, 30, 33). This population also includes adolescents with T1D, who fast against medical advice (9). Individuals with diabetes who fast during Ramadan are more likely to avoid consulting their doctors (12).

 

International Diabetes Federation (IDF) and Diabetes and Ramadan (DAR) International Alliance collaborated to form the IDF-DAR Practical Guidelines 2021 to help healthcare providers (HCPs) better manage diabetes in patients fasting during the month-long holy period of Ramadan (2).

 

It is important to make these individuals with diabetes who cannot fast due to their medical condition understand that they are equally blessed even if they do not fast (30). Many individuals with diabetes who fast prefer to take fasting related advice from their holy leader (Imam). Hence, the right message and education should be disseminated by both the HCPs and the religious leaders (Table 3) (30). HCPs should avoid medical jargons, and counsel patients from a religious standpoint; and religious leaders should integrate into their counseling the value and significance of exemptions in context with the medical advice.

 

Table 3. Medical and Religious Risk Score Recommendations (30)

Risk score

Medical recommendations

Religious recommendations

LOW RISK

0-3 points

Fasting is probably safe. Ensure

1. Medical Evaluation

2. Medication adjustment

3. Strict monitoring

1. Fasting is obligatory

2. Advice not to fast is not allowed except if patient

is unable to fast due to:

3.     -  Physical burden of fasting

4.     -  Has to take medication or food or drink during the fasting hours on medical advice

MODERATE RISK

3.5-6 points

Fasting safety is uncertain

Ensure:

1. Medical Evaluation

2. Medication adjustment

3. Strict monitoring

1. Fasting is preferred but patients may choose

not to fast if they are concerned about their

health after consulting the doctor and taking

into account the full medical circumstances

and patient’s own previous experiences

2. If the patient does fast, they must follow

medical recommendations including regular blood glucose monitoring

HIGH RISK

>6 points

Fasting is probably unsafe

Advise against fasting

 

Medical Nutrition Therapy (MNT) and Ramadan Nutrition Plan (RNP) for People with Diabetes

MNT is an essential component of diabetes management and includes both meal plans and diabetes education, aimed at improving lifestyle and diabetes related behavior (4). MNT helps achieve the desired glycemic control and helps the overweight and obese individuals with T2D improve their lifestyle and lose weight (4). MNT should be appropriate and accurate for the patient’s age, comorbidities, lifestyle requirements, and other medical needs. MNT should be easily absorbed; affordable, easily accessible, acceptable (through right aroma and consider taste preferences), and attractive (visually appealing) (4). This improves adherence to the MNT (4).

 

An MNT plan for individuals with diabetes is essential for safe fasting during Ramadan (32). Structured Ramadan-specific MNT (34) has shown to improve fasting BG and triglyceride levels and pre-dawn and pre-bed SMBG values compared to patients with T2D receiving standard care (34).

 

Structured Ramadan-specific MNT includes (32, 34):

  1. Pre-Ramadan nutrition education
  2. Individualized energy and balanced macronutrient prescriptions for non-fasting period (sunset to sunrise) to prevent hypoglycemia during fasting state
  3. Well distributed carbohydrate intake to prevent post meal hyperglycemia
  4. At least one serving/day of diabetes-specific formula to be taken during Suhoor and/or pre-bed snack.
  5. Diet plan should consider other comorbidities.
  6. Ramadan toolkits:
  • Ramadan flip chart
  • 14-day menu plan
  • Ramadan Nutrition Plate (RNP)
  • Festive season nutrition plan (Syawal nutrition plan)

 

RNP “is a mobile and web-based application designed to help healthcare professionals (HCPs) individualize medical nutrition therapy (MNT) for people with diabetes” who are fasting during Ramadan (32). A well designed and customized RNP is a prerequisite to safe and confident fasting during Ramadan (32). Apart from nutrition, the platform also provides education regarding safe fasting during Ramadan. It helps individuals to safely fast who have no access to HCPs during Ramadan. Several RNPs have been developed for different countries to suit their regional customs, beliefs, and preferences. HCPs can use their country specific RNP, Ramadan Nutrition plate, and well-balanced meal (Table 4) (4, 32) as a guide to individualize the MNT during Ramadan (32).

 

Table 4. Macronutrient Meal Composition for Ramadan (4, 32)

Macronutrient

Recommended amount

Recommended sources

Sources not recommended

Carbohydrate

•                ≤130 g/day

•                Accounts for 40-45% of total caloric intake

•                Adjust as per cultural setting and individual preferences

Low glycemic index and glycemic load carbohydrates: whole grains, legumes, pulses, temperate fruits, green salad, and most vegetables

Foods rich in sugar, refined carbohydrate, processed grains, or starchy foods: sugary beverages, traditional desserts, white rice, white bread, low fiber cereal, and white potatoes

Meal

Calorie%

Carbohydrate exchange*

Suhoor

30-40

3-5

Iftar snack

10-20

1-2

Iftar meal

40-50

3-6

Healthy snack (if required)

10-20

1-2

Fiber

20-35g/day (or 14g /1000 kcal)

High fiber foods: unprocessed food, vegetables, fruits, seeds, pulses, and legumes

 

Fiber helps to provide satiety during Iftar and to delay hunger after Suhoor

-

Protein

•                ≥1.2g/kg of adjusted body weight

•                Accounts for 20-30% of the total caloric intake.

•                Protein enhances satiety and gives sensation of fullness. Also helps to maintain lean body mass

•                Fish, skinless poultry, milk and dairy products, nuts, seeds, and legumes (beans),

•                low fat milk and milk products

•                Protein with a high saturated fat content such as red meat (beef, lamb) and processed meats (increase CVD risk)

Lipids

•                Between 30–35% of the total calorie intake.

•                The type of fat is more important than the total amount of fat in reducing CVD risk.

•                Limit saturated fat to < 7%. PUFA and MUFA should comprise the rest of the fat intake.

• Limit dietary cholesterol to < 300 mg/day or < 200 mg /day if LDL cholesterol > 2.6 mmol/L

•                Consume fat from PUFA and MUFA (e.g., olive oil, vegetable oil, or blended oil (PUFA and Palm oil)). Oily fish (e.g., such as tuna, sardines, salmon, and mackerel) as a source of omega 3-fatty acids

•                Minimize saturated fat, including red meat (beef and lamb), ghee, and foods high in trans-fats (e.g., fast foods, cookies, some margarines).

* 1 Carbohydrate exchange = 15 g Carbohydrates; CVD, cardiovascular disease; MUFA, Monounsaturated fatty acids; PUFA, Polyunsaturated fatty acids

 Medical Management of Diabetes During Ramadan

Diabetes assessment and plan of care during and post Ramadan should ideally follow the flow chart shown in Figure 4 (6,29).

MEDICAL MANAGEMENT OF T1D DURING RAMADAN IN ADOLESCENTS AND YOUNG INDIVIDUALS

TID is treated with insulin replacement therapy. After the Pre-Ramadan risk stratification, adjustments are made to the patient’s dosing, timing, and type of insulin regime based on the patient’s risk level.

Insulin Regimens

There is no conclusive evidence supporting efficacy and safety of a particular insulin regime over another in adolescents with T1D who are fasting during Ramadan. The insulin regime is therefore based on affordability, access to treatment (medication, specialist and advanced technology), and cultural preferences (9).

 

Changing the insulin regime just before Ramdan is likely to result in dose errors and increase the risk of hypoglycemia. Hence, every effort should be made to continue the same regime, but with proper dose modifications and comprehensive counseling covering diet, lifestyle, physical activity, SMBG, and self-titration of insulin dose (10).

 

The most commonly used insulin regimens in adolescents are (9):

  1. Basal-bolus regimens –multiple dose injections (MDI) adjusted according to meal (preferred option)
  2. Conventional twice daily neutral protamine Hagedorn (NPH)/regular short acting (human) insulin
  3. Continuous subcutaneous insulin infusion (CSII) with or without sensors
  4. Premixed insulins (generally not recommended for T1DM)

 

Of these, MDI and CSII are closer to providing the physiological insulin secretion pattern.

Table 5 gives guidance on dose modifications of different insulin regimes.9 SMBG should be encouraged and the patients or their caregivers taught to self-titrate the insulin dose based on the BG levels (Table 6) (10).

 

Table 5. Insulin Dose Adjustments During Ramadan (10,35)

Insulin

Dose modification

Timing

Glucose monitoring

MDI (basal bolus) with analogue insulins

Basal insulin

30-40% dose reduction

Take at Iftar

5–7-point glucose monitoring*

MDI (basal bolus) with analogue insulins

RAI

Suhoor dose reduced 30-50%

Skip pre-lunch dose Iftar dose to be adjusted according to the 2hr post Iftar BG levels

Take at Iftar and Suhoor

5–7-point glucose monitoring*

MDI (basal bolus) with conventional insulins

NPH insulin

No dose modification at Iftar

50% dose reduction at Suhoor

Take at Iftar and Suhoor

5–7-point glucose monitoring or 2-3 staggered readings throughout the day*

MDI (basal bolus) with conventional insulins

Regular insulin

Suhoor dose reduced by 50%

Skip pre-lunch dose

Iftar dose unchanged unless needs to be adjusted according to the 2hr post Iftar BG levels

Take at Iftar and Suhoor

7-point glucose monitoring or 2-3 staggered readings throughout the day*

Premixed (analogue or conventional) once daily

No dose modification

Take at Iftar

At least 2-3 daily

Readings*

Premixed (analogue or conventional) twice daily

No dose modification at Iftar

50% dose reduction at Suhoor

Take at Iftar and Suhoor

At least 2-3 daily

Readings*

CSII / Insulin Pump

Basal rate adjustment

10-30% increase for the initial few hours of Iftar

20-40% decrease for the final 3-4 hours of fast

Bolus doses

Same ICR and ISF principles as followed prior to Ramadan

Reduce the dose post-Suhoor by 20%

CGM

* And whenever any symptoms of hypoglycemia/hyperglycemia develop or feeling unwell

ICR- Insulin Carbohydrate Ratio; ISF- Insulin Sensitivity Factor; RAI- rapid analogue insulin

 

Table 6. SMBG Guided Dose Titrations for Different Types of Insulin During Ramadan (10)

Fasting/pre-Iftar/pre-Suhoor blood glucose

Basal insulin

Short-acting insulin

Premixed insulin

pre-Iftar

pre-Iftar*/post-Suhoor**

pre-Iftar insulin modification

<70 mg/dL (3.9 mmol/L) or symptoms

Reduce by 4 units

Reduce by 4 units

Reduce by 4 units

<90 mg/dL (5.0 mmol/L)

Reduce by 2 units

Reduce by 2 units

Reduce by 2 units

90-126 mg/dL (5.0-7.0 mmol/L)

No change

No change

No change

>126 mg/dL (7.0 mmol/L)

Increase by 2 units

Increase by 2 units

Increase by 2 units

>200 mg/dL (16.7 mmol/L)

Increase by 4 units

Increase by 4 units

Increase by 4 units

*Reduce the insulin dose taken before Suhoor; **Reduce the insulin dose taken before Iftar

Recommendations for Insulin Regimes
  1. T1D management during Ramadan should be individualized according to patient’s need, preference, affordability, acceptability, and access to treatment (9).
  2. The basal-bolus regime is the preferred regime and consists of a long-acting insulin analogue (basal insulin) and a premeal rapid acting insulin analogue (bolus insulin) (9).
  3. Associated with a lower risk of hypoglycemia when compared to conventional, twice-daily, insulin regimens
  4. The bolus insulin dose should be dependent on the carbohydrate count of the meal. It should ideally be given 20 minutes before the meal for better post-prandial BG control.
  5. Boluses covering Suhoor and Iftar should be based on Insulin Carbohydrate Ratio (ICRs) and Insulin Sensitivity Factor (ISFs)

 

Approximately 70% of hypoglycemia occur during the last six hours of fasting. Hence, the type of basal insulin used, reduction in basal insulin dose and modification of insulin timing are the tools used to avoid hypoglycemia:

  • Ramadan fasting should be started with a reduction of basal insulin-starting with 20% and individualizing up to 40% as required
  • Basal insulin can be administered earlier in the day to minimize insulin exposure during the last few hours of fasting when BG levels are low.
  • Basal insulin can also be taken at Iftar or earlier in the evening
  • First-generation basal insulin analogues (such as glargine U-100) are more likely to cause hypoglycemia than the second generation, long-acting insulin analogues (glargine U-300 and degludec). However, the choice of insulin should be individualized based on risk of hypoglycemia.
  • Long-acting insulin analogues are preferred over intermediate acting (NPH/human insulin) as they provide a steady fall of BG towards normal levels by sunset time (9).
  • Twice daily regimens are more likely to be associated with hyperglycemia (9).
  • Twice daily regimens are usually not preferred during Ramadan, but if they are the only choice available to the patient, their timing and dose needs to be more closely monitored depending on the timing, portion size and carbohydrate content of meal (9).
  • Premixed twice daily insulin regimes are not recommended during Ramadan period as they require fixed carbohydrate intake at fixed timing, and this may be difficult for adolescents to follow (9).
  • There is emerging evidence that T1D patients can fast during Ramadan with fewer complications with the help of newer technologies such as insulin pump therapy, CGM and hybrid closed-loop systems (6).
  • CSII with insulin pumps in adolescents help achieve the targeted glycemic control with reduction in hyperglycemia and severe hypoglycemia, and provides more flexibility, improved quality of life and decreased risk of complications like diabetic ketoacidosis (9).
  • CSII allows for easier management of DM and reduces risk of complications than MDI (9)
  • The basal and bolus doses are adjusted through algorithms on the pump or through sensors and mobile applications (in more advanced versions)
  • Basal insulin is reduced by 20-35% in the last 4-5 hours before Iftar and increased by 10-30% after Iftar up to midnight
  • Prandial insulin bolus calculation is based on usual ICR and ISR
  • Bolus doses can be delivered in three different ways:
  • Standard dosing: Immediately before meals
  • Extended or square dosing: gradual dosing over a certain time period
  • Combo or dual wave bolus: combination of standard and extended
  • High fat content diet as seen during Iftar is likely to benefit from extended or combo bolus dosing (9).
  • Insulin pumps augmented with CGM provide better glycemic control and reduce complications considerably in adolescents with T1D. These sensor-augmented pumps are of two types (9):
  • Low Glucose Suspend (LGS) function pumps: The high-risk BG threshold for HE is pre-set in these pumps. The insulin administration can be automatically suspended for ≤2 hours when sensors detect BG levels below the pre-set threshold
  • Predictive Low Glucose Suspend (PLGS) pumps: Insulin administration is automatically suspended before BG reaches hypoglycemic levels (70 mg/dL [3.9 mmol/L]).
  • Automated insulin delivery (closed loop): These can suspend or increase insulin delivery based on sensor-based BG values. Thus, closed loops help increase time in range (TIR) and minimize hypoglycemia and hyperglycemia.
  • Types: Hybrid closed-loop automated insulin delivery systems; Do-It-Yourself Artificial Pancreas Systems (DIY APS)
  • However, CSII is a costly technology, has limited access in many countries, and therefore is not widely available due to cost and accessibility constraints (9).

MEDICAL MANAGEMENT OF ADULTS WITH T1D DURING RAMADAN  

Patients advised to self-monitor BG at 7-time-point points: when fasting; post-breakfast; pre-lunch; post-lunch; pre-dinner; post dinner; and midnight (9).

 

Dose adjustments for the different insulin regimes should start during the pre-Ramadan period and every attempt should be made to attain the desired glycemic goal but at low risk for hypoglycemia.

 

Short acting insulin analogues (glulisine, lispro, or aspart) are associated with less hypoglycemia and better improvement in postprandial glycemia than regular insulin. Premixed insulins are generally not preferred during Ramadan (9).

 

Table 5 provides guidance on dose modifications of different insulin regimes (9). SMBG should be encouraged and the patients taught to self-titrate the insulin dose based on the BG levels (Table 6) (10).

MEDICAL MANAGEMENT OF T2D DURING RAMADAN

Medical management of Ramadan in patients with T2D varies with wide variety of oral and injectable glucose lowering drugs (GLDs) used during Ramadan as shown in Table 9. Patients may be on one or more oral GLDs or a combination of oral and injectable GLDs.

 

Table 9. Different Types of Glucose Lowering Drugs Used by Patients with T2D During Ramadan (1,10)

Oral glucose lowering drugs

Injectable glucose lowering drugs

Sulfonylurea (gliclazide, glipizide, glimepiride, glibenclamide, or glyburide)

Long/intermediate basal insulins (insulin glargine, insulin detemir, insulin degludec or NPH)

 

Insulin: insulin pump, multiple daily injections, insulin lispro, insulin glargine, soluble human insulin, insulin detemir, and biphasic insulin

Biguanides (Metformin)

Bolus prandial rapid or short-acting insulins (lispro, glulisine, aspart or regular human insulin)

Thiazolidinediones (pioglitazone)

Premixed insulins (fixed ratio combinations of short and intermediate acting insulins)-usually not recommended during Ramadan

DPP-4 inhibitors (sitagliptin, saxagliptin, linagliptin, alogliptin, vildagliptin)

GLP-1 RA (lixisenatide, exenatide, liraglutide, dulaglutide, semaglutide)

SGLT2-I (dapagliflozin, canagliflozin, empagliflozin, and ertugliflozin)

Alpha-glucosidase inhibitor (acarbose, voglibose)

Short-acting insulin secretagogues (repaglinide and nateglinide)

Oral GLP-1 RA (semaglutide)

 

  DPP-4, dipeptidyl peptidase 4; GLP-1 RA, Glucagon-like peptide-1 receptor agonists (GLP-1 RAs); NPH, neutral protamine Hagedorn; SGLT2-I, Sodium/glucose cotransporter-2 inhibitors

 

After the Pre-Ramadan risk stratification, adjustments are made to the patient’s GLDs. Some patients may need a change of medications too depending on their risk level. Preference is given to GLDs with better glycemic control and lower risk of hypoglycemia.

Oral Glucose Lowering Drug Adjustments During Ramadan
  • In general, non-sulfonylureas GLDs are superior in lowering hypoglycemia incidence than sulfonylureas (1).
  • Metformin is the most commonly used first line oral GLD, and has minimal risk of hypoglycemia (10).
  • Sulfonylureas are the most commonly used second line oral GLDs after metformin, especially in resource limited settings (10).
  • Short-acting insulin secretagogues can be useful GLDs during Ramadan because of their short duration of action and low risk of hypoglycemia (10).
  • DPP4 inhibitors are well tolerated during fasting and have a low hypoglycemia risk (10).
  • SGLT2 inhibitors are the newest class of oral GLDs used in T2D. They have demonstrated effective glycemic control during Ramadan even in patients with cardiovascular diseases/chronic kidney disease, and have low risk of hypoglycemia (10, 36).
  • An individual should be started on a SGLT2 inhibitor at least 2-4 weeks before Ramadan for the BG levels to stabilize during the fasting time.
  • Of the thiazolidinediones, only pioglitazone is widely approved for T2DM, has low hypoglycemia risk, but clinical data on its use during Ramadan is limited (10).

 

Table 10. Oral Glucose Lowering Drugs Used During Ramadan: Dose Modifications and Timing Adjustments in Individuals with Type 2 Diabetes (10)

Oral GLD

Dose modification

Timing

Metformin once daily

No dose modification

Take at Iftar

Metformin twice daily

No dose modification

Take at Iftar and Suhoor

Metformin thrice daily

No modification to morning dose. Afternoon dose to be combined with evening dose

Take morning dose before Suhoor and evening dose at Iftar

Prolonged release metformin

No dose modification

Take at Iftar

Sulfonylureas once daily

Reduce dose in patients with well controlled BG levels

Take at Iftar

Sulfonylureas twice daily

In patients with well controlled BG levels Iftar dose remains the same. Suhoor dose is reduced

Take at Iftar

Older sulfonylureas (e.g., glibenclamide)

Avoid and replace with 2nd generation SUs such as glicazide, glicazide MR, and glimepiride

Short-acting insulin secretagogues thrice daily dosing

Reduce or re-distribute to two doses

Take before Iftar and Suhoor

DPP4 inhibitor once daily

No dose modification

Take at Iftar

DPP4 inhibitor twice daily (vildagliptin)

No dose modification

Take at Iftar and Suhoor

SGLT2 inhibitors

No dose modification

Take at Iftar

Thiazolidinedione

No dose modification

Take at Iftar

  BG, blood glucose; DPP-4, dipeptidyl peptidase 4; MR, modified release; SGLT2-I, Sodium/glucose cotransporter-2 inhibitors; SU, sulfonylurea

Injectable Glucose Lowering Drug Adjustments During Ramadan
  • Most patients with long-standing T2D eventually need insulin to manage their BG levels. Various insulin regimes are used in T2D (table 9) (1, 10) and in general, the use of insulin increases the risk of hypoglycemia during Ramadan.
  • Insulin can be given as single daily injection, MDI or as CSII through insulin pumps. The insulin regime is therefore based on affordability, access to treatment (medication, specialist and advanced technology), and cultural preferences (9).
  • Changing the insulin regime just before Ramdan is likely to result in dose errors and increase risk of hypoglycemia. Hence, every effort should be made to continue the same regime, but with proper dose modifications and comprehensive counseling covering diet, lifestyle, physical activity, SMBG, and self-titration of insulin dose (10).
  • Table 5 shows the various insulins and how their doses and timing are adjusted during Ramadan.
  • SMBG guided dose titrations for different insulin types are shown in Table 6.
  • GLP-1 RAs can be safely used with other GLDs including metformin and insulin. They have low hypoglycemia risk, but the risk could be higher if given with insulin or sulfonylureas. However, dose needs to be titrated at least 2-4 weeks prior to Ramadan (10).
Individuals on Multiple Antidiabetic Therapy

Individuals on multiple GLDs are at higher risk of hypoglycemia during Ramadan (18). The risk of hypoglycemia is highest if they are on ≥4 GLDs or on a combination of metformin, DPP4I and basal insulin (37).

 

In individuals on multiple GLDs, the risk of hypoglycemia is dependent on several factors such as type and number of GLDs, duration of diabetes, pre-Ramadan glycemic control, renal function, and presence of other comorbidities (10).

 

Individuals on ≥3 GLDs who are fasting during Ramadan should receive comprehensive counseling and advice before the start of Ramadan, and it should cover diet, lifestyle, physical activity, SMBG, and dose and time modifications of GLDs (10).

 

Individuals on a combination of insulin and SUs are at highest risk of hypoglycemia and require a dose reduction GLDs (approximately 25-50% reduction in insulin dose) during Ramadan.

 

Flash glucose monitoring, CGM, activity monitoring, risk stratification, dose adjustments, and use of artificial intelligence-based algorithms that cover one or more of these aspects are the various tools that are likely to help high-risk patients with T2D fast with fewer hypoglycemia and hyperglycemia related complications (6, 10).

MANAGEMENT OF DIABETES IN SPECIAL POPUATIONS DURING RAMADAN

As discussed, individuals who are considered high risk for fasting during Ramadan need special pre-Ramadan risk stratification, counseling, dose modification, and need to follow strict SMBG during Ramadan, and those on insulin should be capable of self-titrating their insulin dose based on their BG values.

 

This is especially true for special population considered high risk due to a high probability of harm caused by fasting during Ramadan (5). This special population of high-risk individuals with diabetes includes pregnant women, elderly, and people with CVD or CKD. All these individuals are usually advised not to fast, but many do decide to fast against medical advice (5).

Management of Diabetes/Gestational Diabetes During Ramadan

Even in healthy pregnant women, fasting during Ramadan results in biochemical changes that almost mimic the effects of prolonged fasting (35). Ramadan fasting results in an increase in triglycerides (TG), free fatty acids (FFA), and ketones in healthy pregnant women along with a decrease in glucose and insulin (35). However, data on physiological and biochemical changes caused by fasting during Ramadan in pregnant women with diabetes is largely lacking.

 

Pregnancy is an exemption from fasting. However, many pregnant women choose to fast during Ramadan. A detailed discussion regarding the potential risks of fasting must be held with them.

While healthy pregnant women can generally fast safely with no maternal or fetal risk, those with hyperglycemia need to strictly monitor their BG levels to prevent hyperglycemia, hypoglycemia, and adverse maternal and fetal outcomes (35).

 

  • Pre-Ramadan assessment should begin months before Ramadan, and apart from risk stratification, should focus on breaking general myths like ‘finger-prick testing for BG levels breaks their fast’, encourage SMBG, and educate about the maternal and fetal risks of both hypoglycemia and hyperglycemia (35).
  • Pregnant women with diabetes should maintain normal physical activity while fasting. The Taraweeh prayer they offer should be considered as exercise for which insulin doses should be adjusted as required (35).
  • Fiber rich food and drinking 2-3 liters of water a day should be encouraged. Suhoor should be taken as late as possible (35).
  • Insulin and/or metformin are the treatment of choice in pregnancy with diabetes. Though glibenclamide is also used in some patients, its use should be discouraged during Ramadan. Some women with gestational diabetes may be managed on diet and/or metformin too.
  • The metformin dose may not need any change during pregnancy but the dose of insulin should be modified as discussed in Table 5 (10, 35). Insulin dose titration should be guided by SMBG as shown in Table 6.
  • SMBG should be carried out as guided in Figure 6. Pregnant women with diabetes should strictly monitor and maintain their BG levels as follows (35):
  • Fasting between 70-95 mg/dL (3.9 – 5.3 mmol/L).
  • Post-prandial < 120 mg/dL (6.7 mmol/L).
  • Pregnant women with diabetes should break their fast if (35):
  • BG levels < 70 mg/dL (3.9 mmol/L) during fasting hours.
  • Feel unwell.
  • Feel reduced fetal movement.
  • Pregnant women with diabetes should carry out regular SMBG at the following time points (35):
  • Before the sunset meal.
  • 1-2 hours after meals
  • Once during the day while fasting, particularly in the afternoon.
  • Anytime they feel unwell.
Management of Diabetes in Elderly with Diabetes Fasting During Ramadan

Older age (≥ 65 years) by itself can be considered a high risk for fasting during Ramadan in individuals with diabetes, even though many elderly fast successfully during Ramadan (38). Older individuals with diabetes are less likely to fast than younger ones (DAR 2020 survey: 71.2% of ≥ 65 years intended to fast compared to 87.3% of those < 65 years) (39). However, fasting during Ramadan being a personal choice, many older adults with diabetes do choose to fast during Ramadan. The DAR Global Survey (2020) also showed that the elderly were more motivated to fast with 69% of those aged ≥ 65 years fasting for 30 days compared to 60% of those < 65 years (39).

 

Elderly (≥ 65 years) with diabetes were significantly more likely to break their fast than younger (<65 years) individuals with diabetes (17% vs. 11.5%; P<0.001) (39). Similarly, they were significantly more likely to break their fast due to hypoglycemia than their younger counterparts (67.7% vs.55.4%; P=0.02).

 

Fasting during Ramadan in elderly with diabetes needs special consideration and attention because:

  • Diabetes related complications are higher in elderly and they need careful BG monitoring and GLD dose adjustments, which should be started well before Ramadan (38).
  • Fasting related complications likely to be seen in elderly with diabetes can be due to both hyperglycemia and hypoglycemia, and also include impaired renal function, impaired postural balance, poor attention, and volume depletion. The risk increases with the number of days fasted (38).
  • The DAR 2020 survey showed that hypoglycemia was significantly higher in elderly as compared to younger population (17.4% vs.15.2%; P<0.001) (39).
  • 9% of those aged ≥ 65 visited the emergency department compared to 4.3% of individuals aged < 65
  • Elderly were also more likely to get hospitalized due to hypoglycemia
  • While 31.5% reduced their GLD dosing, 17% made no change to their medication dose
  • The use of SUs and insulin increases risk of hypoglycemia. 32.7% of elderly need insulin, probably due to long standing diabetes.
  • The DAR 2020 survey also showed that significantly greater number of elderly with diabetes who are fasting had hyperglycemia (BG levels > 16.6 mmol/L or 300 mg/dL) during Ramadan (19.3% vs. 15.6%; P=0.006) (39). 8.4% of the elderly had to attend the emergency department due to hyperglycemia related complications.
  • The DAR 2020 survey showed that the majority (80%) do not break their fast even if they have hyperglycemia, and 20% do not change their behavior (food intake, medication change), 25% reduced their food intake, and 21% increased their medication dose (39).
  • In the DAR 2020 Global Survey, 21% of participants with T2DM aged ≥ 65 years checked their BG levels once or less than once a week. Only around 10% checked their BG levels 3–4 times a day. There was no change in SMBG behavior during Ramadan (39).
  • Research on elderly fasting during Ramadan is largely lacking.38 Landmark trials in Ramadan like the EPIDIAR study which was used to formulate many recommendations for individuals with diabetes fasting during Ramadan, did not include the elderly.7
  • The elderly population is growing fast, and therefore there will be more individuals with diabetes who are ≥ 65 years and intend to fast (40)
  • The risk of fasting is much higher in elderly than in younger population with diabetes (38). This is because the elderly have more comorbidities (hypertension, hyperlipidemia, CVDs, CKD etc.) than the younger population (38, 39).
  • Elderly with diabetes and impaired renal functions, CVD, dementia, frailty, and/or those with risk of falls are at higher risk for complications during fasting than elderly without comorbidities (38). Therefore, risk stratification of elderly with diabetes who decide to fast during Ramadan should be based not only on age, but also on their comorbidities, functional capacity, and ability to manage medications and carry out SMBG, cognition, and social circumstances (38).

 

Hence, the elderly with diabetes are a high-risk category for fasting during Ramadan. They need proper Pre-Ramadan risk stratification, education, and support to ensure that they can fast safely with proper SMBG and medication monitoring.

 

Table 11 covers the basic recommendations for elderly who intend to fast during Ramadan.

 

Table 11. Basic Recommendations for Elderly who Intend to Fast During Ramadan

MEDICATIONS AND REGIMENS

•                Choose medications that have a lower hypoglycemia risk

•                 Make dose adjustments to lower the risk of hypoglycemia

•                 For individuals on SUs, gliclazide and glimepiride should be used instead of glibenclamide

•       SGLT2 inhibitors doses should be reviewed for benefit vs risks of adverse events especially in elderly with impaired renal function or those on diuretics

•       Insulin: dose titration based on SMBG should be taught and dose modifications carried out based on the insulin type

SMBG

•                 Increase frequency to a 5-point time scale

•                 Use CGM if available and feasible

DIET AND PHYSICAL ACTIVITY

•                 Individualized diet and activity plan

•                 Started before Ramadan and adhere during fasting days

•                 Medication doses and timings adjusted according to diet and physical activity level

•                 Adequate nutrition should be stressed and education provided

•                 Hydration ensured through proper planning

SOCIAL SUPPORT

Given that elderly may have cognition, memory, and physical deficits, adequate support should be ensured pre-Ramadan to ensure SMBG, adherence to diet and physical activity plan, insulin dose titration, and oral GLD dose modification

AWARENESS OF RISK OF COMPLICATIONS

•                 Discuss and document symptoms and events to increase awareness and recognition of complications

•                 Both patient and caregiver should be educated to recognize the symptoms of complications

  CGM, continuous glucose monitoring; GLD, glucose lowering drugs; SMBG, self-monitoring of blood glucose

Other Concerns Regarding Management of Diabetes During Ramadan

MANAGEMENT OF COMORBID HYPOTHYROIDISM

Hypothyroidism is commonly seen in patients with diabetes. Usually, thyroxine is taken half an hour before breakfast. However, during Ramadan, the breakfast time is shifted to pre-dawn. This is a time of rush and individuals may find it difficult to time the thyroxine dose half an hour before Suhoor. Similarly, if thyroxine is pushed to evening, then taking it half an hour before Iftar is usually difficult as usually this meal is taken with rest of the family and by Iftar time hunger score is high. Hence, thyroxine may be taken late (after a 4-hour gap) at night as long as no heavy meal is taken between Iftar and late night (12).

BARIATRIC SURGERY

Diabesity (co-existing diabetes and obesity) is of pandemic proportions across the world (4). Bariatric surgery is commonly performed in individuals with diabesity. Bariatric surgery poses certain concerns regarding fasting during Ramadan as these individuals cannot consume large meals and therefore absorb certain macronutrients (12).

GAPS AND WAY FORWARD

The last few decades have contributed immensely to the growing knowledge and clinical experience of health care providers regarding the clinical and metabolic complications of fasting, pre-fasting assessment, risk stratification and initiation of changes in medication dose and timing and dietary/lifestyle modifications during Ramadan (1). However, greater efforts are required to improve communication between the medical experts and religious scholars in order to ensure that medical guidance regarding safe fasting during Ramadan is best received by the public (30). Further well-designed clinical trials are required to assess the best treatment options for adolescents and adults with diabetes who fast during Ramadan. Artificial intelligence, use of RNP and other such tools need to be integrated to ensure safe fasting during Ramadan.

 ACKNOWLEDGEMENTS

All named authors for this manuscript meet the International Committee of Medical Journal Editors (ICMJE) criteria for authorship. All authors take full responsibility for the integrity of the work and have given final approval for the published version. The authors acknowledge Dr. Kokil Mathur and Dr. Punit Srivastava from Mediception Science Pvt. Ltd, Gurgaon, India for providing writing and editing assistance.

REFERENCES  

  1. Abdelrahim D, Faris ME, Hassanein M, et al. Impact of Ramadan Diurnal Intermittent Fasting on Hypoglycemic Events in Patients With Type 2 Diabetes: A Systematic Review of Randomized Controlled Trials and Observational Studies. Frontiers in Endocrinology. 2021;12. Accessed March 2, 2022. https://www.frontiersin.org/article/10.3389/fendo.2021.624423
  2. IDF-DAR Practical Guidelines. DAR practical guidelines for healthcare professionals. Published 2021. Accessed March 2, 2022. https://www.idf.org/our-activities/education/diabetes-and-ramadan/healthcare-professionals.html
  3. Jabbar A, Malek R, Hussein Z. Chapter 2. Epidemiology of diabetes and fasting during Ramadan. In: International Diabetes Federation and DAR International Alliance. Diabetes and Ramadan: Practical Guidelines. International Diabetes Federation and DAR International Alliance; 2021. Accessed March 4, 2022. https://www.idf.org/our-activities/education/diabetes-and-ramadan/healthcare-professionals.html
  4. Kapoor N, Sahay R, Kalra S, et al. Consensus on Medical Nutrition Therapy for Diabesity (CoMeND) in Adults: A South Asian Perspective. Diabetes Metab Syndr Obes. 2021;14:1703-1728. doi:10.2147/DMSO.S278928
  5. Bennakhi A, Buyukbese MA, Al Saleh Y, Almadani AA, Eliana F. Chapter 1. Introduction to the IDF-DAR ractical Guidelines. In: International Diabetes Federation and DAR International Alliance. Diabetes and Ramadan: Practical Guidelines. International Diabetes Federation and DAR International Alliance; 2021. Accessed March 4, 2022. https://www.idf.org/our-activities/education/diabetes-and-ramadan/healthcare-professionals.html
  6. Ahmed SH, Chowdhury TA, Hussain S, et al. Ramadan and Diabetes: A Narrative Review and Practice Update. Diabetes Ther. 2020;11(11):2477-2520. doi:10.1007/s13300-020-00886-y
  7. Salti I, Bénard E, Detournay B, et al. A population-based study of diabetes and its characteristics during the fasting month of Ramadan in 13 countries: results of the epidemiology of diabetes and Ramadan 1422/2001 (EPIDIAR) study. Diabetes Care. 2004;27(10):2306-2311. doi:10.2337/diacare.27.10.2306
  8. Babineaux SM, Toaima D, Boye KS, et al. Multi-country retrospective observational study of the management and outcomes of patients with Type 2 diabetes during Ramadan in 2010 (CREED). Diabet Med. 2015;32(6):819-828. doi:10.1111/dme.12685
  9. Wan Bebakar WM, Wan Mohamad RM, Hafidh K, et al. Chapter 9. Management of Type 1 diabetes when fasting during Ramadan. In: International Diabetes Federation and DAR International Alliance. Diabetes and Ramadan: Practical Guidelines. International Diabetes Federation and DAR International Alliance; 2021. Accessed March 4, 2022. https://www.idf.org/our-activities/education/diabetes-and-ramadan/healthcare-professionals.html
  10. Hanif W, Hassanein M, Elhadd TA, Mohamed NA. Chapter 10. Management of Type 2 diabetes when fasting during Ramadan. In: International Diabetes Federation and DAR International Alliance. Diabetes and Ramadan: Practical Guidelines. International Diabetes Federation and DAR International Alliance; 2021. Accessed March 4, 2022. https://www.idf.org/our-activities/education/diabetes-and-ramadan/healthcare-professionals.html
  11. Lessan N, Ali T. Energy Metabolism and Intermittent Fasting: The Ramadan Perspective. Nutrients. 2019;11(5):E1192. doi:10.3390/nu11051192
  12. Lessan N, Ezzat Faris M, Assaad-Khalil SH, Ali T. Chapter 3. What happens to the body? Physiology of fasting during Ramadan. In: International Diabetes Federation and DAR International Alliance. Diabetes and Ramadan: Practical Guidelines. International Diabetes Federation and DAR International Alliance; 2021. Accessed March 4, 2022. https://www.idf.org/our-activities/education/diabetes-and-ramadan/healthcare-professionals.html
  13. Lessan N, Hannoun Z, Hasan H, Barakat MT. Glucose excursions and glycaemic control during Ramadan fasting in diabetic patients: insights from continuous glucose monitoring (CGM). Diabetes Metab. 2015;41(1):28-36. doi:10.1016/j.diabet.2014.11.004
  14. Al-Rawi N, Madkour M, Jahrami H, et al. Effect of diurnal intermittent fasting during Ramadan on ghrelin, leptin, melatonin, and cortisol levels among overweight and obese subjects: A prospective observational study. PLoS One. 2020;15(8):e0237922. doi:10.1371/journal.pone.0237922
  15. Hassanein M, Al Awadi FF, El Hadidy KES, et al. The characteristics and pattern of care for the type 2 diabetes mellitus population in the MENA region during Ramadan: An international prospective study (DAR-MENA T2DM). Diabetes Res Clin Pract. 2019;151:275-284. doi:10.1016/j.diabres.2019.02.020
  16. Basit A, AlOzairi E, Abdelgadir E. Chapter 4. The effects of fasting during Ramadan on physical and mental wellbeing. In: International Diabetes Federation and DAR International Alliance. Diabetes and Ramadan: Practical Guidelines. International Diabetes Federation and DAR International Alliance; 2021. Accessed March 4, 2022. https://www.idf.org/our-activities/education/diabetes-and-ramadan/healthcare-professionals.html
  17. Husain R, Duncan MT, Cheah SH, Ch’ng SL. Effects of fasting in Ramadan on tropical Asiatic Moslems. Br J Nutr. 1987;58(1):41-48. doi:10.1079/bjn19870067
  18. Jabbar A, Hassanein M, Beshyah SA, Boye KS, Yu M, Babineaux SM. CREED study: Hypoglycaemia during Ramadan in individuals with Type 2 diabetes mellitus from three continents. Diabetes Res Clin Pract. 2017;132:19-26. doi:10.1016/j.diabres.2017.07.014
  19. Ba-Essa EM, Hassanein M, Abdulrhman S, Alkhalifa M, Alsafar Z. Attitude and safety of patients with diabetes observing the Ramadan fast. Diabetes Res Clin Pract. 2019;152:177-182. doi:10.1016/j.diabres.2019.03.031
  20. Mattoo V, Milicevic Z, Malone JK, et al. A comparison of insulin lispro Mix25 and human insulin 30/70 in the treatment of type 2 diabetes during Ramadan. Diabetes Res Clin Pract. 2003;59(2):137-143. doi:10.1016/s0168-8227(02)00202-4
  21. Cesur M, Corapcioglu D, Gursoy A, et al. A comparison of glycemic effects of glimepiride, repaglinide, and insulin glargine in type 2 diabetes mellitus during Ramadan fasting. Diabetes Res Clin Pract. 2007;75(2):141-147. doi:10.1016/j.diabres.2006.05.012
  22. Bakiner O, Ertorer ME, Bozkirli E, Tutuncu NB, Demirag NG. Repaglinide plus single-dose insulin glargine: a safe regimen for low-risk type 2 diabetic patients who insist on fasting in Ramadan. Acta Diabetol. 2009;46(1):63-65. doi:10.1007/s00592-008-0062-7
  23. Hassanein M, Hanif W, Malik W, et al. Comparison of the dipeptidyl peptidase-4 inhibitor vildagliptin and the sulphonylurea gliclazide in combination with metformin, in Muslim patients with type 2 diabetes mellitus fasting during Ramadan: results of the VECTOR study. Curr Med Res Opin. 2011;27(7):1367-1374. doi:10.1185/03007995.2011.579951
  24. Bashier A, Bin Hussain A, MK A. Safety and Efficacy of Liraglutide as an Add-On Therapy to Pre-Existing Anti-Diabetic Regimens during Ramadan, A Prospective Observational Trial. Journal of Diabetes & Metabolism. 2015;06. doi:10.4172/2155-6156.1000590
  25. Hassanein M, Echtay A, Hassoun A, et al. Tolerability of canagliflozin in patients with type 2 diabetes mellitus fasting during Ramadan: Results of the Canagliflozin in Ramadan Tolerance Observational Study (CRATOS). Int J Clin Pract. 2017;71(10). doi:10.1111/ijcp.12991
  26. Aldawi N, Darwiche G, Abusnana S, Elbagir M, Elgzyri T. Initial increase in glucose variability during Ramadan fasting in non-insulin-treated patients with diabetes type 2 using continuous glucose monitoring. Libyan J Med. 2019;14(1):1535747. doi:10.1080/19932820.2018.1535747
  27. Alawadi F, Mohammed K. Bashier A, Rashid F, Chowdhury TA. Chapter 13. Risks of fasting during Ramadan: Cardiovascular, Cerebrovascular and Renal complications. In: International Diabetes Federation and DAR International Alliance. Diabetes and Ramadan: Practical Guidelines. International Diabetes Federation and DAR International Alliance; 2021. Accessed March 4, 2022. https://www.idf.org/our-activities/education/diabetes-and-ramadan/healthcare-professionals.html
  28. Jawad F, Kalra S. Ramadan and diabetes management - The 5 R’s. J Pak Med Assoc. 2015;65(5 Suppl 1):S79-80.
  29. Ahmedani MY, Zainudin SB, AlOzairi E. Chapter 7. Pre-Ramadan Assessment and Education. In: International Diabetes Federation and DAR International Alliance. Diabetes and Ramadan: Practical Guidelines. International Diabetes Federation and DAR International Alliance; 2021. Accessed March 4, 2022. https://www.idf.org/our-activities/education/diabetes-and-ramadan/healthcare-professionals.html
  30. El Sayed AA, Hassanein M, Afandi B, Tayeb K, Diop SN. Chapter 6. Diabetes and Ramadan: A Medico-Religious Perspective. In: International Diabetes Federation and DAR International Alliance. Diabetes and Ramadan: Practical Guidelines. International Diabetes Federation and DAR International Alliance; 2021. Accessed March 4, 2022. https://www.idf.org/our-activities/education/diabetes-and-ramadan/healthcare-professionals.html
  31. Hassanein M, Al-Arouj M, Hamdy O, et al. Diabetes and Ramadan: Practical guidelines. Diabetes Res Clin Pract. 2017;126:303-316. doi:10.1016/j.diabres.2017.03.003
  32. Hamdy O, Mohd Yusof BN, Maher S. Chapter 8. The Ramadan Nutrition Plan (RNP) for people with diabetes. In: International Diabetes Federation and DAR International Alliance. Diabetes and Ramadan: Practical Guidelines. International Diabetes Federation and DAR International Alliance; 2021. Accessed March 4, 2022. https://www.idf.org/our-activities/education/diabetes-and-ramadan/healthcare-professionals.html
  33. Ahmed WN, Arun CS, Koshy TG, et al. Management of diabetes during fasting and COVID-19 – Challenges and solutions. Journal of Family Medicine and Primary Care. 2020;9(8):3797-3806. doi:10.4103/jfmpc.jfmpc_845_20
  34. Mohd Yusof BN, Wan Zukiman WZHH, Abu Zaid Z, et al. Comparison of Structured Nutrition Therapy for Ramadan with Standard Care in Type 2 Diabetes Patients. Nutrients. 2020;12(3):813. doi:10.3390/nu12030813
  35. Afandi B, Hassanein M, Taha Salih B, Abdo S. Chapter 11. Management of hyperglycaemia in pregnancy when fasting during Ramadan. In: International Diabetes Federation and DAR International Alliance. Diabetes and Ramadan: Practical Guidelines. International Diabetes Federation and DAR International Alliance; 2021. Accessed March 4, 2022. https://www.idf.org/our-activities/education/diabetes-and-ramadan/healthcare-professionals.html
  36. Bajaj HS, Abouhassan T, Ahsan MR, et al. Diabetes Canada Position Statement for People With Types 1 and 2 Diabetes Who Fast During Ramadan. Can J Diabetes. 2019;43(1):3-12. doi:10.1016/j.jcjd.2018.04.007
  37. Elhadd T, Dabbous Z, Bashir M, et al. Incidence of hypoglycaemia in patients with type-2 diabetes taking multiple glucose lowering therapies during Ramadan: the PROFAST Ramadan Study. J Diabetes Metab Disord. 2018;17(2):309-314. doi:10.1007/s40200-018-0374-2
  38. Shaltout I, Mohamed M, Iraqi H. Chapter 12. Management of diabetes among the elderly when fasting during Ramadan. In: International Diabetes Federation and DAR International Alliance. Diabetes and Ramadan: Practical Guidelines. International Diabetes Federation and DAR International Alliance; 2021. Accessed March 4, 2022. https://www.idf.org/our-activities/education/diabetes-and-ramadan/healthcare-professionals.html
  39. Hassanein M, Hussein Z, Shaltout I, et al. The DAR 2020 Global survey: Ramadan fasting during COVID 19 pandemic and the impact of older age on fasting among adults with Type 2 diabetes. Diabetes Res Clin Pract. 2021;173:108674. doi:10.1016/j.diabres.2021.108674
  40. IDF. IDF Diabetes Atlas 2021. International Diabetes Federation; 2021. Accessed March 11, 2022. https://diabetesatlas.org/atlas/tenth-edition/

 

Thyroid Nodules and Thyroid Cancer Prior to, During, and Following Pregnancy

ABSTRACT

 

Thyroid cancer is the second most common malignancy to co-occur in pregnancy. Further, the rising prevalence of treated thyroid cancer in women of child-bearing age means that survivors of thyroid cancer are frequently presenting for obstetric care, occasionally in the setting of persisting structural disease. To ensure that optimal health outcomes are achieved for mother and child, it is essential that pre-pregnancy issues are comprehensively addressed, and that management decisions during pregnancy remain both patient and child focused, best achieved through a woman-centered multidisciplinary team. As new data emerge regarding the impact of radioactive iodine on fertility, careful balancing of risk and benefits of this treatment is required. 

 

INTRODUCTION

 

Thyroid nodules are common in women of childbearing age. Thyroid nodules may be detected due to symptoms of local compression (either due to larger size, or pressure on the trachea or esophagus), but are more commonly detected incidentally on imaging performed for other reasons. As well as determining if compressive symptoms are present, all thyroid nodules must be risk-stratified for the presence of malignancy. A third factor is to determine whether the nodule is functional (i.e., autonomously producing thyroid hormone), however this cannot be reliably assessed during pregnancy as it is dependent on radionucleotide imaging which is contra-indicated during pregnancy. 

 

In general, the investigation or treatment of any new or co-existent medical conditions in pregnancy should be weighed against the separate risks and benefits to both the mother and fetus. Although most thyroid nodules will not grow during pregnancy, and therefore permit management decisions to be deferred to after birth (thus prioritizing fetal wellbeing), a small proportion of cases will require emergent management within pregnancy to prioritize maternal wellbeing (1).

 

Thyroid carcinomas generally develop from follicular epithelial cells (termed differentiated thyroid cancer, DTC) and present morphologically as papillary (PTC) or follicular (FTC) subtypes. Anaplastic thyroid cancer (ATC) is a rare, highly aggressive de-differentiated variant of DTC. Rarely, parafollicular, neuro-endocrine derived C-cells can give rise to medullary thyroid cancers (MTC). Thyroid lymphoma and metastases from other solid organ cancers are rare. In general, DTC has an greater than 98% 10 year survival in women of child-bearing age (2).

 

EPIDEMIOLOGY OF THYROID NODULES AND THYROID CANCER 

 

Thyroid Nodules

 

There is a clear female preponderance for the development of thyroid nodules that is demonstrated in studies from varied ethic groups, and in populations that are iodine-replete and iodine deficient (3-5). Thyroid nodules are also more prevalent with increasing age (6){Reiners, 2004 #1161}.  This may partly be explained by exposure to female reproductive hormones, as studies have demonstrated associations with increasing thyroid nodularity and multiparity, older age at menopause, and the presence of uterine fibroids (6-9). In a single study, use of oral contraceptive hormones was associated with reduced thyroid volume, but not a change in thyroid nodularity (10).

 

Carcinoma of the Thyroid Gland

 

The increased prevalence of thyroid nodules in females is matched by an increased prevalence of thyroid cancer amongst women.  SEER data from the United States cancer registry reports thyroid cancer incidence at 21 cases per 100,000 females, compared to 7.1 per 100,000 males (11).  When stratified by age, it is evident that this gender-based divergence is seen as early as puberty (Figure 1). The peak incidence of thyroid cancer amongst females occurs in midlife (age 35-59), and occurs earlier than the peak incidence in males (age 65-75), which corresponds with exposure to female reproductive hormones. ATC and MTC have equal incidence between genders.

Figure 1. Incidence of thyroid cancer by age- and gender- in the United States. Data source: SEER 18 (2010-2014). https://seer.cancer.gov/faststats

Although epidemiological data would suggest a strong link between exposure to female reproductive hormones and development of thyroid cancer, firm evidence linking reproductive factors to thyroid cancer risk is less clear. Some studies have shown a small (or transient) increase risk of DTC following pregnancy compared to nulliparous women (12).  Age of menarche, menopause, and menstrual cycle patterns present conflicting data (12), however in general, longer exposure to reproductive hormones appears associated with increased thyroid cancer risk (13-15). Conversely, extended periods of breastfeeding (resulting in prolonged reductions in cirulating estradiol), have been associated with with decreased incidence of thyroid cancer (14, 16, 17).

 

Incidence of Thyroid Carcinoma in Pregnancy

 

Multiple studies confirm that carcinoma of the thyroid gland is the second most frequent pregnancy-associated cancer, behind carcinoma of the breast. Registry studies suggest that thyroid cancer is present in between 14-27 per 100,000 mothers giving birth (18, 19). In most cases, this represents newly diagnosed thyroid cancer during pregnancy, which is usually organ-confined. However, a combination of increasing diagnosis of thyroid cancer amongst young women and excellent prognosis has resulted in an increasing cohort of survivors of thyroid cancer requiring obstetric care (20). This is demonstrated by data from Taiwan, showing thyroid cancer prevalence amongst women (175 cases per 100,000 women) is 9-fold higher than the incidence (18 cases per 100,000 women) (21). Occasionally, pregnancy occurs in a woman with known or suspected metastatic disease. A recent study from the USA reports that a historical diagnosis of thyroid cancer was the most common cancer present in women presenting for obstetric care (22).

 

IMPACT OF A PREGNANCY ON NUMBER AND SIZE OF THYROID NODULES

 

Impact of Pregnancy Hormones on Thyroid Follicular Epithelium

 

Pregnancy represents a stimulatory environment for thyroid follicular cells. The pregnancy hormone human chorionic gonadotrophin (HCG) is a heterodimeric glycoprotein. Although the beta subunit is unique, the alpha subunit is common to follicle stimulating hormone, luteinizing hormone, and thyroid stimulating hormone (TSH). As a result, this structural homology causes cross-stimulation of the TSH receptor by HCG, leading to physiological TSH-independent stimulation of the TSH-receptor, predominantly in the first trimester when HCG levels are highest. As well as contributing to gestational hyperthyroidism, in this way HCG mediated TSH-receptor signaling acts to stimulate growth of the thyroid follicular epithelium (23). Sustained activation of the signaling cascade mediated by the TSH-receptor has been associated with an increased risk of thyroid cancer in large observational studies. However, it is not known whether more limited periods of increased TSH-receptor signaling, such as would occur during pregnancy, materially contributes to thyroid cancer risk (24).

 

Iodine (not a pregnancy hormone) is a trace element required for normal maternal thyroid function and fetal thyroid development and function. Pregnancy increases maternal demands for iodine and a daily intake of approximately 250-300mcg is recommended (25).  Iodine excess and iodine deficiency states are both associated with an increased prevalence of thyroid nodules (3, 26).  

 

Changes in Number and Size of Thyroid Nodules During Pregnancy

 

As previously outlined, the hormonal environment of pregnancy is associated with the development of new thyroid nodules, and with potential growth of existing thyroid nodules.  Using ultrasound screening, thyroid nodules are demonstrated in 3-21% of pregnant women (26-28), although most nodules are small (<1cm) and not detectable clinically (26).  Prospective studies of pregnant women show an increase in thyroid nodule number and size during pregnancy. In a study of 221 women in China using repeated sonographic evaluation, an increase in nodule volume during pregnancy was shown in 15% of women in whom nodules were already present at baseline evaluation. New thyroid nodules were detected in 13% of the cohort. Post-partum, the number of women with thyroid nodules had increased from 15% to 24% (26). All nodules had a benign sonographic appearance. Similarly, a study of 726 pregnant women in Belgium identified a 3% incidence of thyroid nodules at baseline (determined by two-step screening with palpation followed by ultrasound). Of those with nodules, 60% showed an increase in size of at least 50%. Further, 20% (4/20) of women with regular sonographic surveillance developed new nodules during pregnancy (27).

 

PRESENTATION OF A NEW THYROID NODULE IN THE PREGNANT PATIENT       

 

A thyroid nodule usually comes to attention in pregnancy following the identification of a palpable abnormality. Screening for thyroid nodules in asymptomatic individuals without risk factors, both in the pregnant and non-pregnant population, is not recommended (29).

Thyroid nodules should be assessed using a triple assessment, including clinical assessment, sonographic risk stratification, and biopsy (in selected cases). Scintigraphy, which is part of the standard workup for functional nodules in the non-pregnant population, is contra-indicated in pregnancy due to the risk of ionizing radiation to the fetus.   

 

Important historical factors that increase the chance of a nodule being malignant include the presence of:

 

  • A familial cancer syndrome, including multiple endocrine neoplasia 2 (MEN2), familial PTC, Cowden’s syndrome, familial adenomatosis polyposis, and Carney Complex.
  • Neck irradiation in childhood, e.g., treatment for cancers of the head and neck
  • Exposure to ionizing radiation in early life (age <18 years)

 

On clinical examination, a palpable lump should be characterized. The presence of a large, very firm or rapidly growing nodule should raise concern for malignancy. Neck lymph nodes should be evaluated. Symptoms and signs of compression of local adjacent structures should be sought. 

 

Many thyroid nodules are functional, however the determination of the functional status of a thyroid nodule in pregnancy is limited. Firstly, although TSH should be checked, a low TSH may reflect gestational hyperthyroidism and should be interpreted with reference to the current gestational age. Most functional nodules progress slowly, therefore a pre-pregnancy TSH level which is at, or below, the lower limit of the reference range may provide a helpful clue. Secondly, radioactive isotopes used for thyroid scintigraphy readily cross the placenta, and the radiation exposure to the fetus does not justify the use of this modality in pregnancy.  Therefore, conclusive determination of whether a thyroid nodule is functional (and thus of very low malignant potential), or non-functional, during pregnancy is usually not possible. 

 

Serum biomarkers for thyroid cancer are not currently in routine use.  Although serum calcitonin is highly sensitive for the diagnosis of MTC (30), it is not validated for use in pregnancy, especially as calcitonin levels rise over the course of a normal pregnancy.  Further, its use in assessment of thyroid nodules in non-pregnant women is not universally established. Carcino-embryonic antigen, also a marker of MTC, can rise during pregnancy, and should be interpreted with caution (31).

 

Neck ultrasound is the definitive tool for assessment of thyroid nodules, and is safe in pregnancy. A high-frequency linear transducer is optimal to provide detailed characterization of the sonographic features, including size, echogenicity, shape, margins, the presence of calcification, and the presence of abnormal lymph nodes in the central and lateral neck.  All nodules should be risk stratified according to a validated scoring system, such as from the American Thyroid Association (32) or the American College of Radiology (33).  If fine-needle aspiration biopsy (FNAB) is required, this can be safely performed in all trimesters of pregnancy, with indications identical to that of the non-pregnant population (32). 

 

Nodules with higher risk features, such as larger size, growth in pregnancy, suspicion of extra-thyroidal extension, presence of large-volume nodal metastases, or suspicion for MTC or ATC should be considered for biopsy and surgery during pregnancy.  However, for smaller nodules without any high-risk features, consideration should be given to deferring biopsy (and any planned intervention) until the post-partum period, as several studies have confirmed that there is no survival benefit for surgery during pregnancy for low risk DTC (34).

 

IMPACT OF PREGNANCY ON A NEW DIAGNOSIS OF THYROID CANCER   

 

Impact of Pregnancy on Outcome of Thyroid Cancer

 

A diagnosis of thyroid cancer during pregnancy has the same excellent long term survival outcomes as seen in other settings. Large retrospective studies in the US population between 1962-1999 (35-37) show similar mortality data irrespective of the diagnosis setting (inside or outside pregnancy) or the timing of surgery (during pregnancy or post-partum).  Although these studies have the benefit of long follow up periods, they are inherently retrospective. Further, the ability of these studies to assess impact of pregnancy on thyroid cancer recurrence is limited, not only due to their retrospective nature, but also due to lack of availability of highly sensitive thyroglobulin assays and high resolution neck ultrasound in historic series (34).

 

In contrast, recent studies suggest that thyroid cancer diagnosed in pregnancy may have a higher risk of recurrence. A retrospective study from Italy showed higher rates of persistent or recurrent thyroid cancer in women diagnosed either during pregnancy or within 12 months after birth (60% persistent or recurrent disease), compared to women diagnosed with thyroid cancer more than 12 months after a pregnancy (4% persistent or recurrent disease) or women who were never pregnant (13% persistent or recurrent disease) (38). However, it is important to note that most of the pregnant women with thyroid cancer underwent surgery in the second trimester (73%), and it is possible that a more limited surgical approach in this setting may have confounded these results. 

 

Similarly, a second retrospective Italian study found a higher rate of persistent or recurrent disease in women with thyroid cancer diagnosed within two years of a pregnancy (11%), compared to women diagnosed more than two years after a pregnancy (1%) or those who were never pregnant (5%) (39). 

 

A pathology study from Australia found that DTC diagnosed within 12 months of pregnancy were more likely to be larger, and have nodal metastases than matched controls (40).

 

Overall, these data should reassure clinicians and patients that the impact of pregnancy on a newly diagnosed thyroid cancer is low, with excellent overall survival outcomes. Epidemiological and clinical data would suggest that the stimulatory milieu of pregnancy may contribute to a slightly higher overall risk of recurrence, which should be taken into consideration when planning follow up strategies.  

 

Timing of Thyroidectomy

 

Expert consensus affirms that thyroidectomy can safely be performed in the second trimester, but is often more appropriately deferred to the post-partum period (34). Clinical markers of aggressive pathology, such as large primary size, rapid growth, or bulky lateral neck nodal disease, would support a strategy of earlier surgery. At present, these clinical markers of aggressiveness are detected by specialized thyroid and neck ultrasound, which should occur at first assessment, and subsequently around 20 weeks (to allow for planning of thyroidectomy in the second trimester, if indicated). Suspicion of non-DTC pathology, such as MTC and ATC, should warrant strong consideration of early surgery. 

 

The optimal timing of surgery in the peri-partum period is uncertain.  Whist many women undergo safe surgery and anesthesia in the second trimester, small risks for mother and fetus remain. However, deferring surgery to the post-partum period potentially disrupts dyadic attachment between mother and child, and may interrupt breast-feeding. As evidence is lacking, patient-centered decision making, with inputs from a multi-disciplinary team, is appropriate. 

 

Small case series support a strategy of deferred surgery for low-risk lesions. For example, 19 women with PTC diagnosed around the time of conception were followed sonographically in pregnancy (41). Nearly 70% were microcarcinomas, and 3 cases had sonographic N1 disease. During pregnancy, 3 tumors had a detectable increase in maximal diameter, while 5 increased in volume. In 2 out of 3 with N1 disease, lymph nodes increased in size although no new nodal disease was detected, and the extent of surgery was not changed. Post-partum, 16 cases proceeded to surgery around 12 months following diagnosis.

 

Maternal Supportive Management During Pregnancy

 

Thyroid stimulating hormone (TSH) is a trophic factor for follicular thyroid cells and is associated with progression of thyroid cancer (23). However, there is no evidence to support the practice of pharmacological suppression of TSH to minimize growth of a primary tumor in pregnancy, and exogenous maternal hyperthyroidism is associated with fetal risk.  Maintaining maternal serum TSH within the lower half of the pregnancy-specific reference range is a reasonable therapeutic goal, and levothyroxine should be initiated, if required, to achieve this target.

 

Dietary iodine should not be restricted, as it is essential for fetal thyroid development.  Maternal physiological demands for iodine increase in pregnancy, and maternal iodine deficiency is associated with development of goiter in the mother. 

 

A recent cohort study found that pregnancies complicated by a diagnosis of thyroid cancer prior to or during pregnancy had a higher incidence of venous thromboembolism (odds radio 2.4) and blood transfusions (odds ratio 2.1), however there was no impact on neonatal outcomes (42). Similarly, the rate of post-partum hemorrhage in women with a history of thyroid cancer was higher than controls (odds ratio 1.23) in a large retrospective observational study, however no other adverse maternal, neonatal, or child outcomes (followed to 80 months post-partum) were found (43).

 

Measurement of Serum Thyroglobulin

 

During pregnancy, maternal serum thyroglobulin levels are higher than pre-pregnancy. This may be an effect of stimulation of maternal thyrocytes by estrogen and HCG. Therefore, maternal thyroglobulin levels during pregnancy must be interpreted with caution. Maternal serum thyroglobulin levels return to baseline values within 1-6 months of pregnancy (44-46).  In general, thyroglobulin status should be assessed no earlier than 6 weeks post-partum. 

 

Considerations in the Planning of Radioiodine Therapy

 

Radioactive iodine therapy following total thyroidectomy, either for remnant ablation, or as adjuvant therapy, is recommended for a subset of DTC with higher risk of recurrence (32).  The administration of radioiodine following pregnancy poses unique challenges, both medically and socially. Firstly, radiation safety precautions necessitate that close contact between the mother and her infant (as well as other young children) must be avoided for around 7 days following a radioactive iodine dose (precise recommendations are determined at the time of therapy) (47). Radioactive iodine is contra-indicated in pregnancy, and if administered, the risks to the fetus must be carefully assessed, based on administered dose and gestational age (48). Secondly, breast tissue expresses the sodium-iodide symporter, which is upregulated during lactation to concentrate iodine in breast milk (48, 49).  Consequently, to minimize exposure of breast tissue to ionizing radiation, lactation should cease a minimum of 8 weeks before radioactive iodine and should not be recommenced so as to avoid potential breast-milk associated radioactive iodine exposure. 

 

In light of this, the timing of radioactive iodine (if required) should be considered, balancing the potential risk of DTC progression without treatment, the benefits of a period of breastfeeding, and family unit dynamics. The literature is conflicting as to whether radioiodine administered early (within 3 months) or late (within 12 months), has any impact on prognosis. For example, a large retrospective database study including more than 9,000 patients diagnosed with high-risk PTC (primary tumor >4cm, N1 disease, positive surgical margins) found that timing of radioactive iodine within the first 12 months did not impact mortality (the median survival in this cohort was 75 months), after adjustment for confounders (50). In contrast, a small retrospective study of patients with lower risk DTC (235 cases classified as either ATA Low- or ATA-Intermediate- risk) found that deferring radioactive iodine longer than 3 months post-operatively was associated with higher rates of biochemical incomplete or structural incomplete responses compared to earlier radioactive iodine ablation(19% vs 4%) (51).

 

PRE-CONCEPTION CARE OF WOMEN WITH A HISTORY OF THYROID CANCER             

 

Pregnancy following diagnosis and treatment for thyroid cancer is common, and presents specialized management issues. Nonetheless, excellent obstetric outcomes are expected (52).  Pre-conception counselling is recommended for all women with a past history of thyroid cancer.

 

Checklist: Management issues prior to pregnancy in survivors of thyroid cancer.

 

  1. Assessment of thyroid cancer status:
    • Remission? Assessment of disease status: structural and biochemical
    • Potential impact of pregnancy on disease progression
  2. Impact of prior radioiodine therapy on timing of conception and future fertility
    • Ensure > 6 months between radioactive iodine and conception
  3. Thyroid hormone replacement
    • Pre-pregnancy optimization of levothyroxine replacement
    • Pre-emptive adjustment to levothyroxine dosing following conception
    • Potential for unmasking thyroid hormone insufficiency in women with sufficient pre-pregnancy thyroid hormones from a residual hemithyroid
    • Use of pregnancy supplements that may interfere with levothyroxine absorption

 

Establishment of Thyroid Cancer Status

 

To provide a framework for discussing the potential impact of thyroid cancer on pregnancy, an assessment of disease status is valuable, such as recommended by the ATA in its 2015 guidance (Table 1) (32). Evidently, counselling and management discussions will differ depending on what treatment has previously been received (total thyroidectomy vs hemithyroidectomy), the presence of any functional thyroid hormone production (if prior hemithyroidectomy only), the timing of any radioactive iodine administration, and the presence of any residual cancer. MicroPTC under active surveillance is a distinct management issue which is discussed separately.

 

Table 1. 2015 American Thyroid Association Risk Stratification for DTC

2015 ATA Response-to-Therapy classification

Description

Excellent response

No clinical, biochemical or structural evidence of persistent or recurrent thyroid cancer.

Biochemical-incomplete response

Elevated serum thyroglobulin, or rising anti-thyroglobulin antibodies, in the absence of structural disease identifiable on imaging.

Structural-incomplete response

Persistent or recurrent thyroid cancer visible on imaging, either in neck or distant metastases

Indeterminate response

Non-specific biochemical or structural findings that are not able to be classified as benign or malignant (includes stable/declining anti-thyroglobulin antibody levels without evidence of structural disease)

2015 American Thyroid Association Risk Stratification for DTC, tabulated from Haugen et al. (2016).  Refer to ATA Guideline (32) for full discussion of each class and qualifying criteria (Table 13).

 

In women with a history of MTC, the tumor markers calcitonin and CEA are sensitive to detect residual or recurrent disease, and allow for post-operative risk stratification (53).  There are no studies examining whether pregnancy impacts the prognosis of MTC. 

 

Discussing Impact of Pregnancy on Risk of Recurrence

 

There is a growing body of evidence reporting the long-term oncological outcomes in the setting of pregnancy following treatment for thyroid cancer. Key studies are reviewed below.

 

Leboeuf et al. (46) reported outcomes of 36 women between 1997 and 2006, with pregnancy a median 4 years following treatment for DTC. Three women had structural disease present prior to pregnancy, and of these, one showed growth in a cervical lymph node. A further two women developed recurrence following pregnancy that was not present on pre-operative physical examination. Of the full cohort, 22% had a sustained >20% rise in serum thyroglobulin post-partum. 

 

Rosario et al. (54) describe the outcome of 64 pregnancies, occurring a median of 2.4 years after treatment for DTC. In this cohort, no patient had evidence of structural disease either prior to or following pregnancy. Of the subset 49 women with undetectable thyroglobulin prior to pregnancy, this remained undetectable in the post-partum period. Of the 8 patients with low level thyroglobulin prior to pregnancy, no significant post-partum change was observed. 

 

Hirsch et al. (55) studied the outcome of 63 women, where pregnancy occurred a median of 5 years after treatment for PTC.  Of the subset of 6 women with known structural disease prior to conception, 80% were found to have progressed within 12 months of birth (2 with biochemical progression, 3 with structural progression). Of the subset of 5 women with detectable pre-pregnancy thyroglobulin, no significant change was observed post-partum.  Of the remaining 39 women with undetectable pre-pregnancy thyroglobulin, no progression was observed. 

 

Finally, Rakhlin et al. reported the outcome of pregnancy in 235 women following treatment for DTC (56), retrospectively grouped into ATA Response to Therapy criteria (Table 1).  In the 197 women without structural disease prior to pregnancy, no new structural disease was detected following post-partum evaluation.  However, 8% had a significant rise in thyroglobulin.

 

Overall, these data are reassuring that women with an ATA Excellent response to therapy have a very low risk of DTC progression occurring during pregnancy, and a low risk of DTC progression following pregnancy. As such, additional monitoring of thyroid cancer status during pregnancy for these women is not required (34). 

 

However, women with biochemical or structural evidence of disease may have a progression of their thyroid cancer status as a result of pregnancy.  Based on the above studies, the degree of disease progression appears minor, only affects a subset of women, and does not appear to have an impact on the outcome of the pregnancy. 

 

Reducing the Impacts of Prior Thyroid Cancer Treatment on Pregnancy.

 

LEVOTHYROXINE REPLACEMENT  

 

It is essential that all women who are planning pregnancy receive written instructions for the management of thyroid hormone replacement prior to, and immediately following conception.  Requirements for thyroid hormone rise early in gestation, in part as a result of an increase in thyroid-binding globulin. Adequate levels of thyroid hormones are required for healthy fetal development and pregnancy progression.

 

Women previously treated with hemithyroidectomy may unmask relative thyroid hormone deficiency following conception, and may require early initiation of levothyroxine therapy in the first trimester.

 

Women who have been treated with total thyroidectomy will always require an increase in thyroid hormone replacement at conception, of a magnitude between 15-40% of the total weekly dose. A common practice is to advise women to “double the dose” of levothyroxine that they take on two days of the week as soon as pregnancy is confirmed, with further adjustment based on regular thyroid function tests throughout pregnancy (34, 57). Women who adhered to this advice were more likely to have TSH at the pregnancy target than those that deferred thyroxine adjustment until the first specialist consultation (58). 

 

Importantly, pregnancy multivitamins, iron supplements, or calcium supplements may interfere with the absorption of thyroxine, and women should be specifically instructed to take such supplements at a different time of day to minimize interference (59). 

 

Women should be reassured that levothyroxine is both safe and essential for a healthy pregnancy, as inadvertent discontinuation in early pregnancy has been reported (60). In most cases, the TSH target prior to pregnancy (usually targeting the lower half of the normal range) will remain appropriate in pregnancy. Pharmacological suppression of TSH with supra-physiological doses of levothyroxine could be continued in the setting of persistent structural disease, however care should be taken to avoid overt hyperthyroidism, which increases pregnancy risk. In settings where a TSH-suppression strategy has been pursued outside of pregnancy, but in the absence of known structural disease, a careful balancing of risk and benefit should be considered, as although mild hyperthyroidism in pregnancy has not been shown to lead to maternal or fetal complications, greater degrees of hyperthyroidism are associated with adverse pregnancy outcomes (34, 61, 62).

 

IMPLICATIONS OF PREVIOUS RADIOIODINE  

 

Women should defer conception for at least 6 months after radioactive iodine administration.  This period includes the expected time for radioactive iodine to fully decay (approximately 10 weeks), thus avoiding exposing the fetus to gamma-particle emission). A recent large population-based cohort study found that pregnancy occurring within 5 months of radioactive iodine had a higher rate of congenital malformations (odds ratio 1.74, 95%CI, 1.01-2.97; P = .04), which was not seen if conception occurred after 6 months (63). Deferring pregnancy for at least 6 months has the additional benefit of permitting assessment of the response to radioactive iodine therapy, and to determine that no additional treatment with radioactive iodine would be recommended in the following 15 months (conception, pregnancy and the post-partum period) (64). Stabilization of levothyroxine replacement can also take a period of months.

 

In the 12 months following radioactive iodine, 8-16% of women experience amenorrhea, and 12-31% have menstrual irregularities (65). Several studies (including a meta-analysis) have confirmed a small but significant fall in AMH levels following radioactive iodine, and a slightly earlier age of menopause than women who did not receive radioactive iodine (49.5 vs 51 years) (65, 66). 

 

Most studies have not shown that radioactive iodine has an impact on future fertility (65, 67, 68). However, in a retrospective database study comparing survivors of thyroid cancer, women in the age 35-39 subgroup who received radioactive iodine had a lower birth rate (11 vs 16 births per 1000 woman-years) than women who did not receive radioactive iodine.  However, as the time from diagnosis of thyroid cancer to first live birth was also prolonged in this study, it is not clear whether this finding is due to physician recommendation to delay pregnancy, or the biological effects of radioactive iodine (68). In addition, a recent population case-control study found a higher rate of infertility diagnosis amongst survivors of thyroid cancer (69), however this analysis did not take into account any disease-specific factors such as type of treatment received. 

 

In women with a history of thyroid cancer requiring assisted reproductive techniques, pregnancy outcomes were not different compared to controls, although the number of retrieved oocytes was lower (70). A history of radioactive iodine treatment was not associated with differing rates of clinical pregnancy or live birth rates in this group (71).

 

A large longitudinal study followed 2,673 pregnancies and did not show an increase in maternal or fetal adverse events in women previously administered radioactive iodine (72).  A population-based cohort study of women with thyroid cancer in Korea, comparing 59,483 women who underwent thyroidectomy alone, with 51,976 women who had thyroidectomy followed by radioactive iodine found no difference in pregnancy or obstetric outcomes in the 9.7% of the cohort where pregnancy occurred (63). A further systematic review (67), and meta-analysis (73), pooling additional studies reported similar findings, providing sufficient time had elapsed following radioactive iodine administration. 

 

In men, radioactive iodine may transiently impact testicular function, with a short-term rise in FSH, and decrease in normal sperm morphology seen in prospective studies (74). It is suggested that men avoid fathering children for 4 months following radioactive iodine (75, 76). In men who desire fertility, and who are expected to require high cumulative activities of radioactive iodine, sperm banking should be considered. 

 

Surveillance and Monitoring During Pregnancy

 

Based on available data, women with no structural or biochemical evidence of thyroid cancer do not require DTC-specific monitoring during pregnancy. At present, there is no evidence to guide whether additional post-partum surveillance should be instituted beyond that woman’s current surveillance strategy, however consideration of neck ultrasound and serum thyroglobulin at least 6 months post-partum is reasonable. 

 

For women with ‘ATA Biochemical Incomplete’ or ‘ATA Indeterminate’ classification, surveillance during pregnancy could include periodic neck ultrasound, and determination of thyroglobulin and Tg-Ab levels. Clear evidence of progression of thyroid cancer could prompt an increase in the level of TSH suppression, or rarely prompt expedited delivery. Management in the context of a multidisciplinary team is advised. 

 

CONTINUED ACTIVE SURVELLANCE OF PAPILLARY THRYOID MICROCARCINOMA DURING PREGNANCY

 

Non-operative management of microPTC (<10mm in maximal dimension) is increasing, with emerging data on implications for active surveillance during pregnancy. Shindo et al report 9 women with microPTC followed during pregnancy, finding demonstrable growth in 44% (compared to microPTC growth of 11% in non-pregnant controls) (77). Ito et al reported outcomes of 50 pregnancies with microPTC, finding growth of >1mm in 8%, reduced size in 2%. The remaining 90% of cases showed no growth in pregnancy, and there were no nodal metastases detected (78). Oh et al described 13 microPTC in pregnancy, with a single lesion demonstrating growth (41). The available evidence supports the continuation of active surveillance during pregnancy, monitored with periodic neck ultrasound. However, women contemplating pregnancy who are under active surveillance should be advised that a small number of microPTC will grow during pregnancy, and this may result in anxiety for the patient and clinicians. Further studies are awaited in this population (32).      

 

Germline RET Mutations

 

Women with clinically diagnosed MEN2, or who carry a germline mutation in the REarranged during Transfection (RET) proto-oncogene, should be under the care of a specialized clinical team, and should be offered detailed pre-natal genetic counselling.  Individual RET mutations can be characterized for their risk of early-onset MTC, allowing personalized management decisions. The highest risk mutations should prompt consideration of total thyroidectomy in early childhood (53). The presence of hyperparathyroidism and pheochromocytoma should be biochemically excluded prior to pregnancy in any woman with MEN2.

 

MANAGEMENT OF KNOWN RESIDUAL STRUCTURAL DISEASE IN PREGNANCY

 

Case series of pregnancy in women with co-existent thyroid cancer metastases have been reported. The largest study retrospectively studied a cohort of 124 women from China, aged 16-35 years, with lung metastases from thyroid cancer, stratified by whether pregnancy occurred (n=35) and followed for a median 68-82 months after completing treatment with radioactive iodine (79). This study found that pregnancy after thyroid cancer had no measurable difference in 5 year or 10-year progression free survival or overall survival. 10-year overall survival in the pregnancy group was 86%, compared to 82% in the non-pregnant group. Although the groups appeared to have similar characteristics, it remains possible that women who chose pregnancy had a lower severity of disease than those who avoided pregnancy. 

 

Another study retrospectively described outcomes for 38 women at a large cancer center in the USA (56). Included in the cohort were 10 women with pulmonary metastases at the time of diagnosis (and of whom 7 had persistent structural disease prior to pregnancy). During pregnancy, 29% of women had progression of structural disease (11/38, with 5/38 increasing size of known abnormal nodes, 3/38 with newly abnormal lymph nodes, and 1/38 with progression of distant metastases). In total, 3/38 (~8%) were considered “clinically significant” by the study team (required further treatment within 12 months of birth).

 

These data are reassuring that the clinical impact of pregnancy in the setting of persistent structural disease appears low, despite in vitro studies and smaller case series confirming that pregnancy represents a potentially stimulatory setting for thyroid cancer cells. 

 

In general, TSH suppression should be maintained where benefit is felt to outweigh risk to the pregnancy. Serial neck ultrasound during pregnancy will monitor the status of neck disease, however imaging the chest is usually avoided to minimize ionizing radiation to the chest. Where progression of lung metastases is to be monitored, serial lung function testing may be informative.  

 

Currently approved small molecule tyrosine kinase inhibitors have been shown to have embryotoxicity, fetotoxicity, and teratogenicity in rats and rabbits (80, 81), and pregnancy should be avoided in women on this treatment.  A case report of a pregnancy in a women treated with vandetanib up until 6 weeks gestation described no fetal adverse outcomes (82).

 

REFERENCES

 

  1. Oduncu FS, Kimmig R, Hepp H, Emmerich B. Cancer in pregnancy: maternal-fetal conflict. Journal of cancer research and clinical oncology. 2003;129(3):133-46.
  2. Tuttle RM, Haugen B, Perrier ND. Updated American Joint Committee on Cancer/Tumor-Node-Metastasis Staging System for Differentiated and Anaplastic Thyroid Cancer (Eighth Edition): What Changed and Why? Thyroid. 2017;27(6):751-6.
  3. Song J, Zou SR, Guo CY, Zang JJ, Zhu ZN, Mi M, et al. Prevalence of Thyroid Nodules and Its Relationship with Iodine Status in Shanghai: a Population-based Study. Biomedical and environmental sciences : BES. 2016;29(6):398-407.
  4. Moifo B, Moulion Tapouh JR, Dongmo Fomekong S, Djomou F, Manka'a Wankie E. Ultrasonographic prevalence and characteristics of non-palpable thyroid incidentalomas in a hospital-based population in a sub-Saharan country. BMC medical imaging. 2017;17(1):21.
  5. Knudsen N, Laurberg P, Perrild H, Bulow I, Ovesen L, Jorgensen T. Risk factors for goiter and thyroid nodules. Thyroid. 2002;12(10):879-88.
  6. Reiners C, Wegscheider K, Schicha H, Theissen P, Vaupel R, Wrbitzky R, et al. Prevalence of thyroid disorders in the working population of Germany: ultrasonography screening in 96,278 unselected employees. Thyroid. 2004;14(11):926-32.
  7. Wang K, Yang Y, Wu Y, Chen J, Zhang D, Liu C. The association of menstrual and reproductive factors with thyroid nodules in Chinese women older than 40 years of age. Endocrine. 2015;48(2):603-14.
  8. Spinos N, Terzis G, Crysanthopoulou A, Adonakis G, Markou KB, Vervita V, et al. Increased frequency of thyroid nodules and breast fibroadenomas in women with uterine fibroids. Thyroid. 2007;17(12):1257-9.
  9. Kim MH, Park YR, Lim DJ, Yoon KH, Kang MI, Cha BY, et al. The relationship between thyroid nodules and uterine fibroids. Endocrine journal. 2010;57(7):615-21.
  10. Knudsen N, Bulow I, Laurberg P, Perrild H, Ovesen L, Jorgensen T. Low goitre prevalence among users of oral contraceptives in a population sample of 3712 women. Clin Endocrinol (Oxf). 2002;57(1):71-6.
  11. Thyroid Cancer Bethesda, Maryland: National Cancer Institute; 2016 [Available from: http://seer.cancer.gov/statfacts/html/thyro.html.
  12. Moleti M, Sturniolo G, Di Mauro M, Russo M, Vermiglio F. Female Reproductive Factors and Differentiated Thyroid Cancer. Frontiers in endocrinology. 2017;8:111.
  13. He JL, Zhang C, Hu MJ, Wu HB, Lu XL, Hao JH, et al. Reproductive and menstrual factors for papillary thyroid cancer risk: A case-control study in Chinese women. Cancer Epidemiol. 2021;73:101964.
  14. Kim H, Kim KY, Baek JH, Jung J. Are Pregnancy, Parity, Menstruation and Breastfeeding Risk Factors for Thyroid Cancer? Results from the Korea National Health and Nutrition Examination Survey, 2010 to 2015. Clin Endocrinol (Oxf). 2018.
  15. Schubart JR, Eliassen AH, Schilling A, Goldenberg D. Reproductive Factors and Risk of Thyroid Cancer in Women: An Analysis in the Nurses' Health Study II. Womens Health Issues. 2021;31(5):494-502.
  16. Yi X, Zhu J, Zhu X, Liu GJ, Wu L. Breastfeeding and thyroid cancer risk in women: A dose-response meta-analysis of epidemiological studies. Clinical nutrition (Edinburgh, Scotland). 2016;35(5):1039-46.
  17. Wang M, Gong WW, He QF, Hu RY, Yu M. Menstrual, reproductive and hormonal factors and thyroid cancer: a hospital-based case-control study in China. BMC Womens Health. 2021;21(1):13.
  18. Smith LH, Danielsen B, Allen ME, Cress R. Cancer associated with obstetric delivery: results of linkage with the California cancer registry. American Journal of Obstetrics & Gynecology. 2003;189(4):1128-35.
  19. Cottreau CM, Dashevsky I, Andrade SE, Li DK, Nekhlyudov L, Raebel MA, et al. Pregnancy-Associated Cancer: A U.S. Population-Based Study. J Womens Health (Larchmt). 2019;28(2):250-7.
  20. Lim H, Devesa SS, Sosa JA, Check D, Kitahara CM. Trends in Thyroid Cancer Incidence and Mortality in the United States, 1974-2013. Jama. 2017;317(13):1338-48.
  21. Liu F-C, Lin H-T, Lin S-F, Kuo C-F, Chung T-T, Yu H-P. Nationwide cohort study on the epidemiology and survival outcomes of thyroid cancer. Oncotarget. 2017;8(45):78429-51.
  22. Wu PMMD, Jordan KPP, Chew-Graham CAMMD, Mohamed MOM, Barac AMDP, Lundberg GPMD, et al. In-Hospital Complications in Pregnant Women With Current or Historical Cancer Diagnoses. Mayo Clinic Proceedings. 2021;96(11):2779-92.
  23. Rowe CW, Paul J, Geyde C, Tolosa J, Bendinelli C, McGrath S, et al. Targeting the TSH receptor in thyroid cancer. Endocrine Related Cancer. 2017.
  24. Nieto HR, Boelaert K. Thyroid stimulating hormone in thyroid cancer: does it matter? Endocrine-related cancer. 2016.
  25. Delange F. Iodine requirements during pregnancy, lactation and the neonatal period and indicators of optimal iodine nutrition. Public health nutrition. 2007;10(12a):1571-80; discussion 81-3.
  26. Kung AW, Chau MT, Lao TT, Tam SC, Low LC. The effect of pregnancy on thyroid nodule formation. J Clin Endocrinol Metab. 2002;87(3):1010-4.
  27. Glinoer D, Soto MF, Bourdoux P, Lejeune B, Delange F, Lemone M, et al. Pregnancy in patients with mild thyroid abnormalities: maternal and neonatal repercussions. J Clin Endocrinol Metab. 1991;73(2):421-7.
  28. Struve CW, Haupt S, Ohlen S. Influence of frequency of previous pregnancies on the prevalence of thyroid nodules in women without clinical evidence of thyroid disease. Thyroid. 1993;3(1):7-9.
  29. US Preventative Services Task Force. Screening for thyroid cancer: US Preventive Services Task Force Recommendation Statement. Jama. 2017;317(18):1882-7.
  30. Costante G, Meringolo D, Durante C, Bianchi D, Nocera M, Tumino S, et al. Predictive value of serum calcitonin levels for preoperative diagnosis of medullary thyroid carcinoma in a cohort of 5817 consecutive patients with thyroid nodules. Journal of Clinical Endocrinology and Metabolism. 2007;92(2):450-5.
  31. Ercan S, Kaymaz O, Yucel N, Orcun A. Serum concentrations of CA 125, CA 15-3, CA 19-9 and CEA in normal pregnancy: a longitudinal study. Archives of gynecology and obstetrics. 2012;285(3):579-84.
  32. Haugen BR, Alexander EK, Bible KC, Doherty GM, Mandel SJ, Nikiforov YE, et al. 2015 American Thyroid Association Management Guidelines for Adult Patients with Thyroid Nodules and Differentiated Thyroid Cancer: The American Thyroid Association Guidelines Task Force on Thyroid Nodules and Differentiated Thyroid Cancer. Thyroid. 2016;26(1):1-133.
  33. Tessler FN, Middleton WD, Grant EG, Hoang JK, Berland LL, Teefey SA, et al. ACR Thyroid Imaging, Reporting and Data System (TI-RADS): White Paper of the ACR TI-RADS Committee. Journal of the American College of Radiology : JACR. 2017;14(5):587-95.
  34. Alexander EK, Pearce EN, Brent GA, Brown RS, Chen H, Dosiou C, et al. 2017 Guidelines of the American Thyroid Association for the Diagnosis and Management of Thyroid Disease During Pregnancy and the Postpartum. Thyroid. 2017;27(3):315-89.
  35. Moosa M, Mazzaferri EL. Outcome of differentiated thyroid cancer diagnosed in pregnant women. J Clin Endocrinol Metab. 1997;82(9):2862-6.
  36. Yasmeen S, Cress R, Romano PS, Xing G, Berger-Chen S, Danielsen B, et al. Thyroid cancer in pregnancy. International journal of gynaecology and obstetrics: the official organ of the International Federation of Gynaecology and Obstetrics. 2005;91(1):15-20.
  37. Herzon FS, Morris DM, Segal MN, Rauch G, Parnell T. Coexistent thyroid cancer and pregnancy. Archives of otolaryngology--head & neck surgery. 1994;120(11):1191-3.
  38. Vannucchi G, Perrino M, Rossi S, Colombo C, Vicentini L, Dazzi D, et al. Clinical and molecular features of differentiated thyroid cancer diagnosed during pregnancy. Eur J Endocrinol. 2010;162(1):145-51.
  39. Messuti I, Corvisieri S, Bardesono F, Rapa I, Giorcelli J, Pellerito R, et al. Impact of pregnancy on prognosis of differentiated thyroid cancer: clinical and molecular features. Eur J Endocrinol. 2014;170(5):659-66.
  40. Lee JC, Zhao JT, Clifton-Bligh RJ, Gill AJ, Gundara JS, Ip J, et al. Papillary thyroid carcinoma in pregnancy: a variant of the disease? Ann Surg Oncol. 2012;19(13):4210-6.
  41. Oh H-S, Kim WG, Park S, Kim M, Kwon H, Jeon MJ, et al. Serial Neck Ultrasonographic Evaluation of Changes in Papillary Thyroid Carcinoma During Pregnancy. Thyroid. 2017;27(6):773-7.
  42. Spiegel E, Spence AR, Czuzoj-Shulman N, Abenhaim HA. Pregnancy outcomes after thyroid cancer. J Perinat Med. 2019;47(7):710-6.
  43. Cho GJ, Kim SY, Lee HC, Lee KM, Han SW, Oh MJ, et al. Risk of Adverse Obstetric Outcomes and the Abnormal Growth of Offspring in Women with a History of Thyroid Cancer. Thyroid. 2019;29(6):879-85.
  44. Nakamura S, Sakata S, Komaki T, Kojima N, Kamikubo K, Miyazaki S, et al. Serum thyroglobulin concentration in normal pregnancy. Endocrinologia japonica. 1984;31(6):675-9.
  45. Rasmussen NG, Hornnes PJ, Hegedüs L, Feldt-Rasmussen U. Serum thyroglobulin during the menstrual cycle, during pregnancy, and post partum. Acta Endocrinologica. 1989;121(2):168-73.
  46. Leboeuf R, Emerick LE, Martorella AJ, Tuttle RM. Impact of pregnancy on serum thyroglobulin and detection of recurrent disease shortly after delivery in thyroid cancer survivors. Thyroid. 2007;17(6):543-7.
  47. Sisson JC, Freitas J, McDougall IR, Dauer LT, Hurley JR, Brierley JD, et al. Radiation safety in the treatment of patients with thyroid diseases by radioiodine 131I : practice recommendations of the American Thyroid Association. Thyroid. 2011;21(4):335-46.
  48. Iijima S. Effects of fetal involvement of inadvertent radioactive iodine therapy for the treatment of thyroid diseases during an unsuspected pregnancy. Eur J Obstet Gynecol Reprod Biol. 2021;259:53-9.
  49. Azizi F, Smyth P. Breastfeeding and maternal and infant iodine nutrition. Clinical endocrinology. 2009;70(5):803-9.
  50. Suman P, Wang CH, Abadin SS, Block R, Raghavan V, Moo-Young TA, et al. Timing of radioactive iodine therapy does not impact overall survival in high-risk papillary thyroid carcinoma. Endocr Pract. 2016;22(7):822-31.
  51. Li H, Zhang YQ, Wang C, Zhang X, Li X, Lin YS. Delayed Initial Radioiodine Therapy Related to Incomplete Response in Low- to Intermediate-Risk Differentiated Thyroid Cancer. Clin Endocrinol (Oxf). 2018.
  52. Liu D, Wei Y, Zhao Y, Li R, Yan J, Qiao J. Obstetric outcomes in thyroid cancer survivors: A retrospective cohort study. International journal of gynaecology and obstetrics: the official organ of the International Federation of Gynaecology and Obstetrics. 2021;155(1):119-24.
  53. Wells SA, Jr., Asa SL, Dralle H, Elisei R, Evans DB, Gagel RF, et al. Revised American Thyroid Association guidelines for the management of medullary thyroid carcinoma. Thyroid. 2015;25(6):567-610.
  54. Rosario PW, Barroso AL, Purisch S. The effect of subsequent pregnancy on patients with thyroid carcinoma apparently free of the disease. Thyroid. 2007;17(11):1175-6.
  55. Hirsch D, Levy S, Tsvetov G, Weinstein R, Lifshitz A, Singer J, et al. Impact of pregnancy on outcome and prognosis of survivors of papillary thyroid cancer. Thyroid. 2010;20(10):1179-85.
  56. Rakhlin L, Fish S, Tuttle RM. Response to Therapy Status Is an Excellent Predictor of Pregnancy-Associated Structural Disease Progression in Patients Previously Treated for Differentiated Thyroid Cancer. Thyroid. 2017;27(3):396-401.
  57. Alexander EK, Marqusee E, Lawrence J, Jarolim P, Fischer GA, Larsen PR. Timing and magnitude of increases in levothyroxine requirements during pregnancy in women with hypothyroidism. The New England journal of medicine. 2004;351(3):241-9.
  58. Minakata M, Ito M, Kishi T, Hada M, Masaki Y, Nakamura T, et al. The magnitude of increased Levothyroxine dose during pregnancy in patients on thyroid-stimulating hormone (TSH) suppression treatment after total thyroidectomy for papillary carcinoma. Endocrine journal. 2021.
  59. Zamfirescu I, Carlson HE. Absorption of Levothyroxine When Coadministered with Various Calcium Formulations. Thyroid. 2011;21(5):483-6.
  60. Rowe C, Murray K, Woods A, Gupta S, Smith R, Wynne K. Metastatic Thyroid Cancer in Pregnancy: Risk and Uncertainty. Endocrinolgy Diabetes & Metabolism Case Reports. 2016.
  61. Casey BM, Dashe JS, Wells CE, McIntire DD, Leveno KJ, Cunningham FG. Subclinical hyperthyroidism and pregnancy outcomes. Obstetrics and gynecology. 2006;107(2 Pt 1):337-41.
  62. Millar LK, Wing DA, Leung AS, Koonings PP, Montoro MN, Mestman JH. Low birth weight and preeclampsia in pregnancies complicated by hyperthyroidism. Obstetrics and gynecology. 1994;84(6):946-9.
  63. Kim HO, Lee K, Lee SM, Seo GH. Association Between Pregnancy Outcomes and Radioactive Iodine Treatment After Thyroidectomy Among Women With Thyroid Cancer. JAMA Intern Med. 2020;180(1):54-61.
  64. International Atomic Energy Agency. Radiation protection of pregnant women in nuclear medicine: International Atomic Energy Agency;[Available from: https://www.iaea.org/resources/rpop/health-professionals/nuclear-medicine/pregnant-women.
  65. Piek MW, Postma EL, van Leeuwaarde R, de Boer JP, Bos AME, Lok C, et al. The Effect of Radioactive Iodine Therapy on Ovarian Function and Fertility in Female Thyroid Cancer Patients: A Systematic Review and Meta-Analysis. Thyroid. 2021;31(4):658-68.
  66. Yaish I, Azem F, Gutfeld O, Silman Z, Serebro M, Sharon O, et al. A Single Radioactive Iodine Treatment Has a Deleterious Effect on Ovarian Reserve in Women with Thyroid Cancer: Results of a Prospective Pilot Study. Thyroid. 2018;28(4):522-7.
  67. Sawka AM, Lakra DC, Lea J, Alshehri B, Tsang RW, Brierley JD, et al. A systematic review examining the effects of therapeutic radioactive iodine on ovarian function and future pregnancy in female thyroid cancer survivors. Clin Endocrinol (Oxf). 2008;69(3):479-90.
  68. Wu JX, Young S, Ro K, Li N, Leung AM, Chiu HK, et al. Reproductive outcomes and nononcologic complications after radioactive iodine ablation for well-differentiated thyroid cancer. Thyroid. 2015;25(1):133-8.
  69. Velez MP, Richardson H, Baxter NN, McClintock C, Greenblatt E, Barr R, et al. Risk of infertility in female adolescents and young adults with cancer: a population-based cohort study. Hum Reprod. 2021;36(7):1981-8.
  70. Huang N, Zeng L, Yan J, Chi H, Qiao J. Analysis of in vitro fertilization/intracytoplasmic sperm injection outcomes in infertile women with a history of thyroid cancer: a retrospective study. Reproductive Biology and Endocrinology. 2021;19(1):82.
  71. Huang N, Zeng L, Yan J, Chi H, Qiao J. Impact of thyroid cancer treatment on assisted reproductive technology outcomes in women with infertility. J Assist Reprod Genet. 2021;38(8):2121-8.
  72. Garsi JP, Schlumberger M, Rubino C, Ricard M, Labbe M, Ceccarelli C, et al. Therapeutic administration of 131I for differentiated thyroid cancer: radiation dose to ovaries and outcome of pregnancies. J Nucl Med. 2008;49(5):845-52.
  73. Zhang L, Huang Y, Zheng Y, Cai L, Wen J, Chen G. The effect of I-131 therapy on pregnancy outcomes after thyroidectomy in patients with differentiated thyroid carcinoma: a meta-analysis. Endocrine. 2021;73(2):301-7.
  74. Bourcigaux N, Rubino C, Berthaud I, Toubert ME, Donadille B, Leenhardt L, et al. Impact on testicular function of a single ablative activity of 3.7 GBq radioactive iodine for differentiated thyroid carcinoma. Hum Reprod. 2018;33(8):1408-16.
  75. Pacini F, Gasperi M, Fugazzola L, Ceccarelli C, Lippi F, Centoni R, et al. Testicular function in patients with differentiated thyroid carcinoma treated with radioiodine. J Nucl Med. 1994;35(9):1418-22.
  76. Poppe K, Bisschop P, Fugazzola L, Minziori G, Unuane D, Weghofer A. 2021 European Thyroid Association Guideline on Thyroid Disorders prior to and during Assisted Reproduction. Eur Thyroid J. 2021;9(6):281-95.
  77. Shindo H, Amino N, Ito Y, Kihara M, Kobayashi K, Miya A, et al. Papillary thyroid microcarcinoma might progress during pregnancy. Thyroid. 2014;24(5):840-4.
  78. Ito Y, Miyauchi A, Kudo T, Ota H, Yoshioka K, Oda H, et al. Effects of Pregnancy on Papillary Microcarcinomas of the Thyroid Re-Evaluated in the Entire Patient Series at Kuma Hospital. Thyroid. 2015;26(1):156-60.
  79. Xi C, Zhang Q, Song H-J, Shen C-T, Zhang G-Q, Sun J-W, et al. Pregnancy Does not Affect the Prognoses of Differentiated Thyroid Cancer Patients With Lung Metastases. The Journal of Clinical Endocrinology & Metabolism. 2021;106(8):e3185-e97.
  80. U.S Food & Drug Administration. Lenvima Prescribing Information: FDA; 2015 [Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/208692s000lbl.pdf.
  81. U.S Food & Drug Administration. Nexavar Prescribing Information: FDA; 2013 [Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2013/021923s016lbl.pdf.
  82. Thomas N, Glod J, Derse-Anthony C, Baple EL, Obsborne N, Sturley R, et al. Pregnancy on vandetanib in metastatic medullary thyroid carcinoma associated with multiple endocrine neoplasia type 2B. Clin Endocrinol (Oxf). 2018.

Skin Manifestations of Diabetes Mellitus

ABSTRACT

 

Diabetes mellitus is a common and debilitating disease that affects a variety of organs including the skin. Between thirty and seventy percent of patients with diabetes mellitus, both type 1 and type 2, will present with a cutaneous complication of diabetes mellitus at some point during their lifetime. A variety of dermatologic manifestations have been linked with diabetes mellitus; these conditions vary in severity and can be benign, deforming, and even life-threatening. Such skin changes can offer insight into patients’ glycemic control and may be the first sign of metabolic derangement in undiagnosed patients with diabetes. Recognition and management of these conditions is important in maximizing the quality of life and in avoiding serious adverse effects in patients with diabetes mellitus.

 

INTRODUCTION

 

The changes associated with diabetes mellitus can affect multiple organ systems. Between thirty and seventy percent of patients with diabetes mellitus, both type 1 and type 2, will present with a cutaneous complication of diabetes mellitus at some point during their lifetime (1). Dermatologic manifestations of diabetes mellitus have various health implications ranging from those that are aesthetically concerning to those that may be life-threatening. Awareness of cutaneous manifestations of diabetes mellitus can provide insight into the present or prior metabolic status of patients. The recognition of such findings may aid in the diagnosis of diabetes, or may be followed as a marker of glycemic control. The text that follows describes the relationship between diabetes mellitus and the skin, more specifically: (1) skin manifestations strongly associated with diabetes, (2) non-specific dermatologic signs and symptoms associated with diabetes, (3) dermatologic diseases associated with diabetes, (4) common skin infections in diabetes, and (5) cutaneous changes associated with diabetes medications.

 

SKIN MANIFESTATIONS STRONGLY ASSOCIATED WITH DIABETES MELLITUS

 

Acanthosis Nigricans

 

EPIDEMIOLOGY

 

Acanthosis nigricans (AN) is a classic dermatologic manifestation of diabetes mellitus that affects men and women of all ages. AN is more common in type 2 diabetes mellitus (2) and is more prevalent in those with darker-skin color. AN is disproportionately represented in African Americans, Hispanics, and Native Americans (3). AN is observed in a variety of endocrinopathies associated with resistance to insulin such as acromegaly, Cushing syndrome, obesity, polycystic ovarian syndrome, and thyroid dysfunction. Unrelated to insulin resistance, AN can also be associated with malignancies such as gastric adenocarcinomas and other carcinomas (4).

 

PRESENTATION

 

AN presents chronically as multiple poorly demarcated plaques with grey to dark-brown hyperpigmentation and a thickened velvety to verrucous texture. Classically, AN has a symmetrical distribution and is located in intertriginous or flexural surfaces such as the back of the neck, axilla, elbows, palmer hands (also known as “tripe palms”), inframammary creases, umbilicus, or groin. Affected areas are asymptomatic; however, extensive involvement may cause discomfort or fetor. Microscopy shows hyperkeratosis and epidermal papillomatosis with acanthosis. The changes in skin pigmentation are primarily a consequence of hyperkeratosis, not changes in melanin. AN can present prior to the clinical diagnosis of diabetes; the presence of AN should prompt evaluation for diabetes mellitus and for other signs of insulin resistance.

 

PATHOGENESIS

 

The pathogenesis of AN is not completely understood. The predominant theory is that a hyperinsulin state activates insulin growth factor receptors (IGF), specifically IGF-1, on keratinocytes and fibroblasts, provoking cell proliferation, resulting in the aforementioned cutaneous manifestations of AN (5,6).

 

TREATMENT

 

Treatment of AN may improve current lesions and prevent future cutaneous manifestations. AN is best managed with lifestyle changes such as dietary modifications, increased physical activity, and weight reduction. In patients with diabetes, pharmacologic adjuvants, such as metformin, that improve glycemic control and reduce insulin resistance are also beneficial (7). Primary dermatologic therapies are usually ineffective especially in patients with generalized involvement. However, in those with thickened or macerated areas of skin, oral retinoids or topical keratolytics such as ammonium lactate, retinoic acid, or salicylic acid can be used to alleviate symptoms (8-10).

 

Diabetic Dermopathy

 

EPIDEMIOLOGY

 

Dermopathy (DD), also known as pigmented pretibial patches or diabetic shin spots, is the most common dermatologic manifestations of diabetes, presenting in as many as one-half of those with diabetes (11). Although disputed, some consider the presence of DD to be pathognomonic for diabetes. DD has a strong predilection for men and those older than 50 years of age (12). Although DD may antecede the onset of diabetes, it occurs more frequently as a late complication of diabetes and in those with microvascular disease. Nephropathy, neuropathy, and retinopathy are regularly present in patients with DD. An association with cardiovascular disease has also been identified, with one study showing 53% of non-insulin-dependent diabetes mellitus with DD had coexisting coronary artery disease (13).

 

PRESENTATION

 

DD initially presents with rounded, dull, red papules that progressively evolve over one-to-two weeks into well-circumscribed, atrophic, brown macules with a fine scale (figure 1). Normally after about eighteen to twenty-four months, lesions dissipate and leave behind an area of concavity and hyperpigmentation. At any time, different lesions can present at different stages of evolution. The lesions are normally distributed bilaterally and localized over bony prominences. The pretibial area is most commonly involved, although other bony prominences such as the forearms, lateral malleoli or thighs may also be involved. Aside from the aforementioned changes, patients are otherwise asymptomatic. DD is a clinical diagnosis that should not require a skin biopsy. Histologically, DD is rather nonspecific; it is characterized by lymphocytic infiltrates surrounding vasculature, engorged blood vessels in the papillary dermis, and dispersed hemosiderin deposits. Moreover, the histology varies based on the stage of the lesion. Immature lesions present with epidermal edema as opposed to epidermal atrophy which is representative of older lesions (14).

Figure 1. Diabetic Dermopathy

PATHOGENESIS

 

The origin of DD remains unclear, however, mild trauma to affected areas (15), hemosiderin and melanin deposition (16), microangiopathic changes (17), and destruction of subcutaneous nerves (18) have all been suggested.

 

TREATMENT

 

Treatment is typically avoided given the asymptomatic and self-resolving nature of DD as well as the ineffectiveness of available treatments. However, DD often occurs in the context of microvascular complications and neuropathies (12); hence, patients need to be examined and followed more rigorously for these complications. Although it is important to manage diabetes and its complications accordingly, there is no evidence that improved glycemic control alters the development of DD.

 

Diabetic Foot Syndrome

 

EPIDEMIOLOGY

 

Diabetic Foot Syndrome (DFS) encompasses the neuropathic and vasculopathic complications that develop in the feet of patients with diabetes. Although preventable, DFS is a significant cause of morbidity, mortality, hospitalization, and reduction in quality of life of patients with diabetes. The incidence and prevalence of DFS in patients with diabetes is 1% to 4% and 4% to 10%, respectively (19). Furthermore, DFS is slightly more prevalent in type 1 diabetes compared with type 2 diabetes (20). A more comprehensive review of diabetic foot syndrome can be found in The Diabetic Foot chapter of Endotext.

 

PRESENTATION

 

DFS presents initially with callosities and dry skin related to diabetic neuropathy. In later stages, chronic ulcers and a variety of other malformations of the feet develop. Between 15% and 25% of patients with diabetes will develop ulcers (21). Ulcers may be neuropathic, ischemic, or mixed. The most common type of ulcers are neuropathic ulcers, a painless ulceration resulting from peripheral neuropathy. Ulcers associated with peripheral vascular ischemia are painful but less common. Ulcers tend to occur in areas prone to trauma, classically presenting at the site of calluses or over bony prominences. It is common for ulcers to occur on the toes, forefoot, and ankles. Untreated ulcers usually heal within one year, however, fifty percent of patients with diabetes will have recurrence of the ulcer within three years (22). The skin of affected patients, especially in those with type 2 diabetes, is more prone to fungal infection and the toe webs are a common port of entry for fungi which can then infect and complicate ulcers (23). Secondary infection of ulcers is a serious complication that can result in gangrenous necrosis, osteomyelitis, and may even require lower extremity amputation. Another complication, diabetic neuro-osteoarthropathy (also known as Charcot foot), is an irreversible debilitating and deforming condition involving progressive destruction of weight-bearing bones and joints. Diabetic neuro-osteoarthropathy occurs most frequently in the feet and can result in collapse of the midfoot, referred to as “rocker-bottom foot.” Moreover, a reduction of the intrinsic muscle volume and thickening of the plantar aponeurosis can cause a muscular imbalance that produces a clawing deformation of the toes. An additional complication of diabetes and neuropathy involving the feet is erythromelalgia. Erythromelalgia presents with redness, warmth, and a burning pain involving the lower extremities, most often the feet. Symptoms may worsen in patients with erythromelalgia with exercise or heat exposure and may improve with cooling (24).

 

PATHOGENESIS

 

The pathogenesis of DFS involves a combination of inciting factors that coexist together: neuropathy (25), atherosclerosis (25), and impaired wound healing (26). In the setting of long-standing hyperglycemia, there is an increase in advanced glycosylation end products, proinflammatory factors, and oxidative stress which results in the demyelination of nerves and subsequent neuropathy (27,28). Single-cell RNA sequencing revealed that there is a unique subset of fibroblasts that overexpress factors associated with healing within the wound bed as opposed to the wound edge (28A). Additionally, wound healers demonstrate an increase in M1 macrophages as opposed to non-wound healers which have an increase in M2 macrophages. The effect on sensory and motor nerves, can blunt the perception of adverse stimuli and produce an altered gait, increasing the likelihood of developing foot ulcers and malformations. Also damage to autonomic nerve fibers causes a reduction in sweating which may leave skin in the lower extremity dehydrated and prone to fissures and secondary infection (29). In addition to neuropathy, accelerated arterial atherosclerosis can lead to peripheral ischemia and ulceration (30). It has been reported that diabetic patients with Charcot neuroarthropathy are associated with greater impairment of cutaneous microvascular reactivity when compared to non-complicated diabetic groups (30A). Finally, hyperglycemia impairs macrophage functionality as well as increases and prolongs the inflammatory response, slowing the healing of ulcers (31).

 

TREATMENT

 

Treatment should involve an interdisciplinary team-based approach with a focus on prevention and management of current ulcers. Prevention entails daily surveillance, appropriate foot hygiene, and proper footwear, walkers, or other devices to minimize and distribute pressure. An appropriate wound care program should be used to care for ongoing ulcers. Different classes of wound dressing should be considered based on the wound type. Hydrogels, hyperbaric oxygen therapy, topical growth factors, and biofabricated skin grafts are also available (19). The clinical presentation should indicate whether antibiotic therapy or wound debridement is necessary (19). In patients with chronic treatment resistant ulcers, underlying ischemia should be considered; these patients may require surgical revascularization or bypass.

 

Diabetic Thick Skin

 

Skin thickening is frequently observed in patients with diabetes. Affected areas of skin can appear thickened, waxy, or edematous. These patients are often asymptomatic but can have a reduction in sensation and pain. Although different parts of the body can be involved, the hands and feet are most frequently involved. Ultrasound evaluation of the skin can be diagnostic and exhibit thickened skin. Subclinical generalized skin thickening is the most common type of skin thickening. Diabetic thick skin may represent another manifestation of scleroderma-like skin changes or limited joint mobility, which are each described in more detail below.

 

Scleroderma-Like Skin Changes

 

EPIDEMIOLOGY

 

Scleroderma-like skin changes are a distinct and easily overlooked group of findings that are commonly observed in patients with diabetes. Ten to fifty percent of patients with diabetes present with the associated skin findings (32). Scleroderma-like skin changes occurs more commonly in those with type 1 diabetes and in those with longstanding disease (33). There is no known variation in prevalence between males and females, or between racial groups.

 

PRESENTATION

 

Scleroderma-like skin changes develop slowly and present with painless, indurated, occasionally waxy appearing, thickened skin. These changes occur symmetrically and bilaterally in acral areas. In patients with scleroderma-like skin changes the acral areas are involved, specifically the dorsum of the fingers (sclerodactyly), proximal interphalangeal, and metacarpophalangeal joints. Severe disease may extend centrally from the hands to the arms or back. A small number of patients with diabetes may develop more extensive disease, which presents earlier and with truncal involvement. The risk of developing nephropathy and retinopathy is increased in those with scleroderma-like skin changes who also have type 1 diabetes (33,34). The aforementioned symptoms are also associated with diabetic hand syndrome which may present with limited joint mobility, palmar fibromatosis (Dupuytren's contracture), and stenosing tenosynovitis (“trigger finger”) (35). The physical exam finding known as the “prayer sign” (inability to flushly press palmar surfaces on each hand together) may be present in patients with diabetic hand syndrome and scleroderma-like skin changes (36). On histology, scleroderma-like skin changes reveal thickening of the dermis, minimal-to-absent mucin, and increased interlinking of collagen. Although on physical exam scleroderma may be difficult to distinguish from these skin changes, scleroderma-like skin changes are not associated with atrophy of the dermis, Raynaud’s syndrome, pain, or telangiectasias.

 

PATHOGENESIS

 

Although not fully understood, the pathogenesis is believed to involve the strengthening of collagen as a result of reactions associated with advanced glycosylation end products or a buildup of sugar alcohols in the upper dermis (37,38).

 

TREATMENT

 

Scleroderma-like skin changes is a chronic condition that is also associated with joint and microvascular complication. Therapeutic options are extremely limited. One observational report has suggested that very tight blood sugar control may result in the narrowing of thickened skin (39). In addition, aldose reductase inhibitors, which limit increases in sugar alcohols, may be efficacious (38). In patients with restricted ranges of motions, physical therapy can help to maintain and improve joint mobility.

 

Limited Joint Mobility

 

Limited Joint Mobility (LJM), also known as diabetic cheiroarthropathy, is a relatively common complication of long-standing diabetes mellitus. The majority of patients with LJM also present with scleroderma-like skin changes (38,40). The prevalence of LJM is 4% to 26% in patients without diabetes and 8% to 58% in patients with diabetes (41). LJM presents with progressive flexed contractures and hindered joint extension, most commonly involving the metacarpophalangeal and interphalangeal joints of the hand. The earliest changes often begin in the joints of the fifth finger before then spreading to involve the other joints of the hand (38). Patients may present with an inability to flushly press the palmar surfaces of each of their hands together (“prayer sign”) (figure 2) or against the surface of a table when their forearms are perpendicular to the surface of the table (“tabletop sign”) (42). These changes occur as a result of periarticular enlargement of connective tissue. The pathogenesis likely involves hyperglycemia induced formation of advanced glycation end-products, which accumulate to promote inflammation and the formation of stiffening cross-links between collagen (43). LJM is strongly associated with microvascular and macrovascular changes and diagnosis of LJM should prompt a workup for related sequela (44). Patients with LJM may also be at increased risk for falls (45). There are no curative treatments. Symptomatic patients may benefit from non-steroidal anti-inflammatory drugs or targeted injection of corticosteroids (43). LJM is best managed with improved glycemic control (46), as well as, regular stretching to maintain and minimize further limitations in joint mobility.

 

Figure 2. Limited Joint Mobility

Scleredema Diabetocorum

 

EPIDEMIOLOGY

 

Scleredema diabeticorum is a chronic and slowly progressive sclerotic skin disorder that is often seen in the context of diabetes. Whereas 2.5% to 14% of all patients with diabetes have scleredema, over 50% of those with scleredema present with concomitant diabetes (47). Scleredema has a proclivity for those with a long history of diabetes. It remains unclear whether there is a predilection for scleredema in those with type 1 diabetes (48) compared to those with type 2 diabetes (48). Women are affected more often than men (49). Although all ages are affected, scleredema occurs more frequently in those over the age of twenty (48,49).

 

PRESENTATION

 

Scleredema presents with gradually worsening indurated and thickened skin. These skin changes occur symmetrically and diffusely. The most commonly involved areas are the upper back, shoulders, and back of the neck. The face, chest, abdomen, buttocks, and thighs may also be involved; however, the distal extremities are classically spared. The affected areas are normally asymptomatic but there can be reduced sensation. Patients with severe longstanding disease may develop a reduced range of motion, most often affecting the trunk. In extreme cases, this can lead to restrictive respiratory problems. A full thickness skin biopsy may be useful in supporting a clinical presentation. The histology of scleredema displays increased collagen and a thickened reticular dermis, with a surrounding mucinous infiltrate, without edema or sclerosis.

 

PATHOGENESIS

 

Although many theories center on abnormalities in collagen, there is no a consensus regarding the pathogenesis of scleredema. The pathogenesis of scleredema may involve an interplay between non-enzymatic glycosylation of collagen, increased fibroblast production of collagen, or decreases in collagen breakdown (50,51).

 

TREATMENT

 

Scleredema diabeticorum is normally unresolving and slowly progressive over years. Improved glycemic control may be an important means of prevention but evidence has not shown clinical improvements in those already affected by scleredema diabeticorum. A variety of therapeutic options have been proposed with variable efficacy. Some of these therapies include immunosuppressants, corticosteroids, intravenous immunoglobulin, and electron-beam therapy (52). Phototherapy with UVA1 or PUVA may be effective in those that are severely affected (52). Independent of other treatments, physical therapy is an important therapeutic modality for patients with scleredema and reduced mobility (53).

 

Necrobiosis Lipoidica

 

EPIDEMIOLOGY

 

Necrobiosis lipoidica (NL) is a rare chronic granulomatous dermatologic disease that is seen most frequently in patients with diabetes. Although nearly one in four patients presenting with NL will also have diabetes, less than 1% of patients with diabetes will develop NL (54). For unknown reasons, NL expresses a strong predilection for women compared to men (55). NL generally occurs in type 1 diabetes during the third decade of life, as opposed to type 2 diabetes in which it commonly presents in the fourth or fifth decades of life (54). The majority of cases of NL presents years after a diagnosis of diabetes mellitus; however, 14% to 24% of cases of NL may occur prior to or at the time of diagnosis (56). One study evaluating comorbidities and diabetic complications in patients with NL found high rates of smoking, hypertension, hyperlipidemia, obesity, coronary artery disease, myocardial infarction, thyroid disease, poor kidney function, and poor glucose control (56A). The highest comorbidity rates in patients with NL were patients with type 2 diabetes.

 

PRESENTATION

 

NL begins as a single or group of firm well-demarcated rounded erythematous papules (figure 3). The papules then expand and aggregate into plaques characterized by circumferential red-brown borders and a firm yellow-brown waxen atrophic center containing telangiectasias. NL occurs bilaterally and exhibits Koebnerization. Lesions are almost always found on the pretibial areas of the lower extremities. Additional involvement of the forearm, scalp, distal upper extremities, face, or abdomen may be present on occasion, and the heel of the foot or glans penis even more infrequently. If left untreated, only about 15% of lesions will resolve within twelve years. Despite the pronounced appearance of the lesions, NL is often asymptomatic. However, there may be pruritus and hypoesthesia of affected areas, and pain may be present in the context of ulceration. Ulceration occurs in about one-third of lesions, and has been associated with secondary infections and squamous cell carcinoma. The histology of NL primarily involves the dermis and is marked by palisading granulomatous inflammation, necrobiotic collagen, a mixed inflammatory infiltrate, blood vessel wall thickening, and reduced mucin.

Figure 3. Necrobiosis Lipoidica

PATHOGENESIS

 

The pathogenesis of NL is not well understood. The relationship between diabetes and NL has led some to theorize that diabetes-related microangiopathy is related to the development of NL (54). Other theories focus on irregularities in collagen, autoimmune disease, neutrophil chemotaxis, or blood vessels (57).

 

TREATMENT

 

NL is a chronic, disfiguring condition that can be debilitating for patients and difficult for clinicians to manage. Differing degrees of success have been reported with a variety of treatments; however, the majority of such reports are limited by inconsistent treatment responses in patients and a lack of large controlled studies. Corticosteroids are often used in the management of NL and may be administered topically, intralesionally, or orally. Corticosteroids can be used to manage active lesions, but is best not used in areas that are atrophic. Success has also been reported with calcineurin inhibitors (e.g., cyclosporine), anti-tumor necrosis factor inhibitors (e.g., infliximab), pentoxifylline, antimalarials (e.g., hydroxychloroquine), PUVA, granulocyte colony stimulating factor, dipyridamole, and low-dose aspirin (54). Appropriate wound care is important for ulcerated lesions; this often includes topical antibiotics, protecting areas vulnerable to injury, emollients, and compression bandaging. Surgical excision of ulcers typically has poor results. Some ulcerated lesion may improve with split-skin grafting. Although still recommended, improved control of diabetes has not been found to lead to an improvement in skin lesions. Patients with newly diagnosed NL should be screened for hypertension, hyperlipidemia, and thyroid disease (56A). 

Bullosis Diabeticorum

 

EPIDEMIOLOGY

 

Bullosis diabeticorum (BD) is an uncommon eruptive blistering condition that presents in those with diabetes mellitus. Although BD can occasionally present in early-diabetes (58), it often occurs in long-standing diabetes along with other complications such as neuropathy, nephropathy, and retinopathy. In the United States, the prevalence of BD is around 0.5% amongst patients with diabetes and is believed to be higher in those with type 1 diabetes (13). BD is significantly more common in male patients than in female patients (59). The average age of onset is between 50 and 70 years of age (59).

 

PRESENTATION

 

BD presents at sites of previously healthy-appearing skin with the abrupt onset of one or more non-erythematous, firm, sterile bullae. Shortly after forming, bullae increase in size and become more flaccid, ranging in size from about 0.5 cm to 5 cm. Bullae frequently present bilaterally involving the acral areas of the lower extremities. However, involvement of the upper extremities and even more rarely the trunk can be seen. The bullae and the adjacent areas are nontender. BD often presents acutely, classically overnight, with no history of trauma to the affected area. Generally, the bullae heal within two to six weeks, but then commonly reoccur. Histological findings are often non-specific but are useful in distinguishing BD from other bullous diseases. Histology, typically shows an intraepidermal or subepidermal blister, spongiosis, no acantholysis, minimal inflammatory infiltrate, and normal immunofluorescence.

 

PATHOGENESIS

 

There is an incomplete understanding of the underlying pathogenesis of BD and no consensus regarding a leading theory. Various mechanisms have been proposed, some of which focus on autoimmune processes, exposure to ultraviolet light, variations in blood glucose, neuropathy, or changes in microvasculature (60).

 

TREATMENT

 

BD resolve without treatment and are therefore managed by avoiding secondary infection and the corresponding sequelae (e.g., necrosis, osteomyelitis). This involves protection of the affected skin, leaving blisters intact (except for large blisters, which may be aspirated to prevent rupture), and monitoring for infection. Topical antibiotics are not necessary unless specifically indicated, such as with secondary infection or positive culture.

NONSPECIFIC DERMATOLOGIC SIGNS AND SYMPTOMS

Ichthyosiform Changes of the Shins

 

Ichthyosiform changes of the shins presents with large bilateral areas of dryness and scaling (sometimes described as “fish scale” skin) (figure 4). Although cutaneous changes may occur on the hands or feet, the anterior shin is most classically involved. These cutaneous changes are related to rapid skin aging and adhesion defects in the stratum corneum (61). The prevalence of ichthyosiform changes of the shins in those with type 1 diabetes has been reported to be between 22% to 48% (33,62). These changes present relatively early in the disease course of diabetes. There is no known difference in prevalence between males and females (33). The development of ichthyosiform changes of the shins is related to production of advanced glycosylation end products and microangiopathic changes. Treatment is limited but topical emollients or keratolytic agents may be beneficial (61).

Figure 4. Acquired ichthyosiform changes

Xerosis

 

Xerosis is one of the most common skin presentations in patients with diabetes and has been reported to be present in as many as 40% of patients with diabetes (63). Xerosis refers to skin that is abnormally dry. Affected skin may present with scaling, cracks, or a rough texture. These skin changes are most frequently located on the feet of patients with diabetes. It has been reported that diabetic patients that are obese will experience more severe hypohidrosis of the feet (63A). In patients with diabetes, xerosis occurs often in the context of microvascular complications (40). To avoid complications such as fissures and secondary infections, xerosis can be managed with emollients like ammonium lactate (64).

 

Acquired Perforating Dermatosis

 

EPIDEMIOLOGY

 

Perforating dermatoses refers to a broad group of chronic skin disorders characterized by a loss of dermal connective tissue. A subset of perforating dermatoses, known as acquired perforating dermatoses (APD), encompasses those perforating dermatoses that are associated with systemic diseases. Although APD may be seen with any systemic diseases, it is classically observed in patients with chronic renal failure or long-standing diabetes (65). APD occurs most often in adulthood in patients between the ages of 30 and 90 years of age (65,66). The prevalence of APD is unknown. It is estimated that of those diagnosed with APD about 15% also have diabetes mellitus (67). In a review, 4.5% to 10% of patients with chronic renal failure presented with concurrent APD (68,69).

 

PRESENTATION

 

APD presents as groups of hyperkeratotic umbilicated-nodules and papules with centralized keratin plugs. The lesions undergo Koebnerization and hence the extensor surfaces of the arms and more commonly the legs are often involved; eruptions also occur frequently on the trunk. However, lesions can develop anywhere on the body. Lesions are extremely pruritic and are aggravated by excoriation. Eruptions may improve after a few months but an area of hyperpigmentation typically remains. Histologically, perforating dermatoses are characterized by a lymphocytic infiltrate, an absence or degeneration of dermal connective tissue components (e.g., collagen, elastic fibers), and transepidermal extrusion of keratotic material.

 

PATHOGENESIS

 

The underlying pathogenesis is disputed and not fully understood. It has been suggested that repetitive superficial trauma from chronic scratching may induce epidermal or dermal derangements (70). The glycosylation of microvasculature or dermal components has been suggested as well. Other hypotheses have implicated additional metabolic disturbances, or the accumulation of unknown immunogenic substances that are not eliminated by dialysis (65). APD is also considered a form of prurigo nodularis (70A).

 

TREATMENT

 

APD can be challenging to treat and many of the interventions have variable efficacy. Minimizing scratching and other traumas to involved areas can allow lesions to resolve over a period of months. This is best achieved with symptomatic relief of pruritus. Individual lesions can be managed with topical agents such as keratolytics (e.g., 5% to 7% salicylic acid), retinoids (e.g., 0.01% to 0.1% tretinoin), or high-potency steroids (71). Refractory lesions may respond to intralesional steroid injections or cryotherapy (71). A common initial approach is a topical steroid in combination with emollients and an oral antihistamine. Generalized symptoms may improve with systemic therapy with oral retinoids, psoralen plus UVA light (PUVA), allopurinol (100 mg daily for 2 to 4 months), or oral antibiotics (doxycycline or clindamycin) (72). Additionally, as APD is a form of prurigo nodularis, the use of immunomodulating agents such as dupilumab may be effective in treating the condition. There is evidence of dupilumab monotherapy effectively treating certain forms of APD (72A). Nevertheless, effective management of the underlying systemic disease is fundamental to the treatment of APD. In those with diabetes, APD is unlikely to improve without improved blood sugar control. Moreover, dialysis does not reduce symptoms; however, renal transplantation can result in the improvement and resolution of cutaneous lesions.

 

Eruptive Xanthomas

 

EPIDEMIOLOGY

 

Eruptive xanthomas (EX) is a clinical presentation of hypertriglyceridemia, generally associated with serum triglycerides above 2,000 mg/dL (73). However, in patients with diabetes, lower levels of triglycerides may be associated with EX. The prevalence of EX is around one percent in type 1 diabetes and two percent in type 2 diabetes (74,75). Serum lipid abnormalities are present in about seventy-five percent of patients with diabetes (76).

 

PRESENTATION

 

EX has been reported as the first presenting sign of diabetes mellitus, granting it can present at any time in the disease course. EX presents as eruptions of clusters of glossy pink-to-yellow papules, ranging in diameter from 1 mm to 4 mm, overlying an erythematous area (figure 5). The lesions can be found on extensor surfaces of the extremities, the buttocks, and in areas susceptible in Koebnerization. EX is usually asymptomatic but may be pruritic or tender. The histology reveals a mixed inflammatory infiltrate of the dermis which includes triglyceride containing macrophages, also referred to as foam cells.

Figure 5. Eruptive Xanthomas

PATHOGENESIS

 

Lipoprotein lipase, a key enzyme in the metabolism of triglyceride rich lipoproteins, is stimulated by insulin. In an insulin deficient state, such as poorly controlled diabetes, there is decreased lipoprotein lipase activity resulting in the accumulation of chylomicrons and other triglyceride rich lipoproteins (77). Increased levels of these substances are scavenged by macrophages (78). These lipid-laden macrophages then collect in the dermis of the skin where they can lead to eruptive xanthomas.

 

TREATMENT

 

EX can resolve with improved glycemic control and a reduction in serum triglyceride levels (79). This may be achieved with fibrates or omega-3-fatty acids in addition to an appropriate insulin regimen (80). A more comprehensive review of the treatment of hypertriglyceridemia can be found in the Triglyceride Lowering Drugs section of Endotext.

 

Acrochordons

 

Acrochordons (also known as soft benign fibromas, fibroepithlial polyps, or skin tags) are benign, soft, pedunculated growths that vary in size and can occur singularly or in groups (figure 6). The neck, axilla, and periorbital area, are most frequently involved, although other intertriginous areas can also be affected. Skin tags are common in the general population, but are more prevalent in those with increased weight or age, and in women. It has been reported that as many as three out of four patients presenting with acrochordons also have diabetes mellitus (81). Patients with acanthosis nigricans may have acrochordons overlying the affected areas of skin. Although disputed, some studies have suggested that the amount of skin tags on an individual may correspond with an individual's risk of diabetes or insulin resistance (82). Excision or cryotherapy is not medically indicated but may be considered in those with symptomatic or cosmetically displeasing lesions.

Figure 6. Acrochordons

Diabetes-Associated Pruritus

 

Diabetes can be associated with pruritus, more often localized than generalized. Affected areas can include the scalp, ankles, feet, trunk, or genitalia (83,84). Pruritus is more likely in patients with diabetes who have dry skin or diabetic neuropathy. Specifically, for type 2 diabetes, risk factors for pruritus were identified to be age, duration of disease, diabetic peripheral neuropathy, diabetic retinopathy, diabetic chronic kidney disease, and fasting plasma glucose levels (84A). Involvement of the genitalia or intertriginous areas may occur in the setting of infection (e.g., candidiasis). Treatments include topical capsaicin, topical ketamine-amitriptyline-lidocaine, oral anticonvulsants (e.g., gabapentin or pregabalin), and, in the case of candida infection, antifungals.

 

Huntley’s Papules (Finger Pebbles)

 

Huntley’s papules, also known as finger pebbles, are a benign cutaneous finding affecting the hands. Patients present with clusters of non-erythematous, asymptomatic, small papules on the dorsal surface of the hand, specifically affecting the metacarpophalangeal joints and periungual areas. The clusters of small papules can develop into coalescent plaques. Other associated cutaneous findings include hypopigmentation and induration of the skin. Huntley’s papules are strongly associated with type 2 diabetes and may be an early sign of diabetic thick skin (85,86). Topical therapies are usually ineffective; however, patients suffering from excessive dryness of the skin may benefit from 12% ammonium lactate cream (87).

 

Keratosis Pilaris

 

Keratosis pilaris is a very common benign keratotic disorder. Patients with keratosis pilaris classically present with areas of keratotic perifollicular papules with surrounding erythema or hyperpigmentation (figure 7). The posterior surfaces of the upper arms are often affected but involvement of the thighs, face, and buttocks can also be seen. Compared to the general population, keratosis pilaris occurs more frequently and with more extensive involvement of the skin in those with diabetes (33,62). Keratosis pilaris can be treated with various topical therapies, including salicylic acid, moisturizers, and emollients.

Figure 7. Keratosis Pilaris

Pigmented Purpuric Dermatoses

 

Pigmented purpuric dermatoses (also known as pigmented purpura) are associated with diabetes, more often in the elderly, and frequently coexists with diabetic dermopathy (88,89). Pigmented purpura presents with non-blanching copper-colored patches involving the pretibial areas of the legs or the dorsum of the feet. The lesions are usually asymptomatic but may be pruritic. Pigmented purpuric dermatoses occur more often in late-stage diabetes in patients with nephropathy and retinopathy as a result of microangiopathic damage to capillaries and sequential erythrocyte deposition (90).

 

Palmar Erythema

 

Palmar erythema is a benign finding that presents with symmetric redness and warmth involving the palms. The erythema is asymptomatic and often most heavily affects the hypothenar and thenar eminences of the palms. The microvascular complications of diabetes are thought to be involved in the pathogenesis of palmar erythema (91). Although diabetes associated palmar erythema is distinct from physiologic mottled skin, it is similar to other types of palmar erythema such as those related to pregnancy and rheumatoid arthritis.

 

Periungual Telangiectasias

 

As many as one in every two patients with diabetes are affected by periungual telangiectasias (92). Periungual telangiectasias presents asymptomatically with erythema and telangiectasias surrounding the proximal nail folds (71). Such findings may occur in association with “ragged” cuticles and fingertip tenderness. The cutaneous findings are due to venous capillary dilatation that occurs secondary to diabetic microangiopathy. Capillary abnormalities, such as venous capillary tortuosity, may differ and can represent an early manifestation of diabetes-related microangiopathy (93).

 

Rubeosis Faciei

 

Rubeosis faciei is a benign finding present in about 7% of patients with diabetes; however, in hospitalized patients, the prevalence may exceed 50% (94). Rubeosis faciei presents with chronic erythema of the face or neck. Telangiectasias may also be visible. The flushed appearance is often more prominent in those with lighter colored skin. The flushed appearance is thought to occur secondary to small vessel dilation and microangiopathic changes. Complications of diabetes mellitus, such as retinopathy, neuropathy, and nephropathy are also associated with rubeosis faciei (90). Facial erythema may improve with better glycemic control and reduction of caffeine or alcohol intake.

 

Yellow Skin and Nails

 

It is common for patients with diabetes, particularly elderly patients with type 2 diabetes, to present with asymptomatic yellow discolorations of their skin or fingernails. These benign changes commonly involve the palms, soles, face, or the distal nail of the first toe. The accumulation of various substances (e.g., carotene, glycosylated proteins) in patients with diabetes may be responsible for the changes in complexion; however, the pathogenesis remains controversial (95).

 

Onychocryptosis

 

Onychocryptosis, or ingrown toenails, have been reported in patients with diabetes, specifically type 2 diabetes (95A). The great toes are most affected. It is hypothesized that this nail change can occur in diabetic patients because onychocryptosis is correlated with increased body mass index, trauma, weak vascular supply, nail plate dysfunction, and subungual hyperkeratosis.

 

DERMATOLOGIC DISEASES ASSOCIATED WITH DIABETES

 

Generalized Granuloma Annulare

 

EPIDEMIOLOGY

 

Although various forms of granuloma annulare exist, only generalized granuloma annulare (GGA) is thought to be associated with diabetes. It is estimated that between ten and fifteen percent of cases of GGA occur in patients with diabetes (96). Meanwhile, less than one percent of patients with diabetes present with GGA. GGA occurs around the average age of 50 years. It occurs more frequently in women than in men, and in those with type 1 diabetes (97).

 

PRESENTATION

 

GGA initially presents with groups of skin-colored or reddish, firm papules which slowly grow and centrally involute to then form hypo- or hyper-pigmented annular rings with elevated circumferential borders. The lesions can range in size from 0.5 cm to 5.0 cm. The trunk and extremities are classically involved in a bilateral distribution. GGA is normally asymptomatic but can present with pruritus. The histology shows dermal granulomatous inflammation surrounding foci of necrotic collagen and mucin. Necrobiosis lipoidica can present similarly to GGA; GGA is distinguished from necrobiosis lipoidica by its red color, the absence of an atrophic epidermis, and on histopathology: the presence of mucin and lack of plasma cells.

 

PATHOGENESIS

 

The pathogenesis of GGA is incompletely understood. It is believed to involve an unknown stimulus that leads to the activation of lymphocytes through a delayed-type hypersensitivity reaction, ultimately initiating a proinflammatory cascade and granuloma formation (98).

 

TREATMENT

 

GGA has a prolonged often unresolving disease course and multiple treatments have been suggested to better manage GGA. However, much of the information stems from small studies and case reports. Antimalarials, retinoids, corticosteroids, dapsone, cyclosporine, PUVA, and calcineurin inhibitors have been suggested as therapies (98).

 

Psoriasis

 

Psoriasis is a chronic immune-mediated inflammatory disorder that may present with a variety of symptoms, including erythematous, indurated, and scaly areas of skin. Psoriasis has been found to be associated with a variety of risk factors, such as hypertension and metabolic syndrome, that increase the likelihood of cardiovascular disease. The development of diabetes mellitus, an additional cardiovascular risk factor, has been strongly associated with psoriasis (99). In particular, younger patients and those with severe psoriasis may be more likely to develop diabetes in the future (99)

 

Lichen Planus

 

Lichen planus is a mucocutaneous inflammatory disorder characterized by firm, erythematous, polygonal, pruritic, papules. These papules classically involve the wrists or ankles, although the trunk, back, and thighs can also be affected. A number of studies have cited an association between lichen planus and abnormalities in glucose tolerance testing. Approximately one in four patients with lichen planus have diabetes mellitus (100). Although the association is contested, it has been reported that patients with diabetes may also be more likely to develop oral lichen planus (101).

 

Vitiligo

 

Vitiligo is an acquired autoimmune disorder involving melanocyte destruction. Patients with vitiligo present with scattered well-demarcated areas of depigmentation that can occur anywhere on the body, but frequently involves the acral surfaces and the face. Whereas about 1% of the general population is affected by vitiligo, vitiligo is much more prevalent in those with diabetes mellitus. Vitiligo occurs more frequently in women and is also more common in type 1 than in type 2 diabetes mellitus (96,98). Coinciding vitiligo and type 1 diabetes mellitus may be associated with endocrine autoimmune abnormalities of the gastric parietal cells, adrenal, or thyroid (102). A more comprehensive review of polyglandular autoimmune disorders can be found in the Autoimmune Polyglandular Syndromes section of Endotext.

 

Hidradenitis Supparativa

 

Hidradenitis supparativa (HS) is a chronic inflammatory condition characterized by inflamed nodules and abscesses located in intreginious areas such as the axilla or groin. These lesions are often painful and malodorous. HS is frequently complicated by sinus formation and the development of disfiguring scars. HS occurs more often in women than men and usually presents in patients beginning in their twenties (103). Compared to the general population, diabetes mellitus is three-times more common in patients with HS (104). It is recommended that patients with HS be screened for diabetes mellitus. There is no standardized approach to the treatment of HS, although some benefits have been reported with the use of antibiotics, retinoids, antiandrogens, or immunomodulators such as tumor necrosis factor (TNF) inhibitors (105).

 

Glucagonoma

 

Glucagonoma is a rare neuroendocrine tumor that most frequently affects patients in their sixth decade of life (106). Patients with glucagonoma may present with a variety of non-specific symptoms. However, necrolytic migratory erythema (NME) is classically associated with glucagonoma and presents in 70% to 83% of patients (106) (107). NME is characterized by erythematous erosive crusted or vesicular eruptions of papules or plaques with irregular borders. The lesions may become bullous or blistered, and may be painful or pruritic. The abdomen, groin, genitals, or buttocks are frequently involved, although cheilitis or glossitis may also be present. Biopsy at the edge of the lesion may demonstrate epidermal pallor, necrolytic edema, and a perivascular inflammatory infiltrate (108). Patients with glucagonoma may also present with diabetes mellitus. In patients with glucagonoma, diabetes mellitus frequently presents prior to NME (107). Approximately 20% to 40% of patients will present with diabetes mellitus before the diagnosis of glucagonoma (107,109). Of those patients diagnosed with glucagonoma but not diabetes mellitus, 76% to 94% will eventually develop diabetes mellitus (110). A more comprehensive review of glucagonoma can be found in the Glucagonoma section of Endotext.

 

Skin Infections

 

The prevalence of cutaneous infections in patients with diabetes is about one in every five patients (111). Compared with the general population, patients with diabetes mellitus are more susceptible to infections and more prone to repeated infections. A variety of factors are believed to be involved in the vulnerability to infection in patients with uncontrolled diabetes, some of these factors include angiopathy, neuropathy, hindrance of the anti-oxidant system, abnormalities in leukocyte adherence, chemotaxis, and phagocytosis, as well as, a glucose-rich environment facilitates the growth of pathogens.

 

BACTERIAL

 

Erysipelas and cellulitis are cutaneous infections that occur frequently in patients with diabetes. Erysipelas presents with pain and well-demarcated superficial erythema. Cellulitis is a deeper cutaneous infection that presents with pain and poorly-demarcated erythema. Folliculitis is common among patients with diabetes, and is characterized by inflamed, perifollicular, papules and pustules. Treatment for the aforementioned conditions depends on the severity of the infection. Uncomplicated cellulitis and erysipelas are typically treated empirically with oral antibiotics, whereas uncomplicated folliculitis may be managed with topical antibiotics. Colonization with methicillin-resistant Staphylococcus aureus (MRSA) is not uncommon among patients with diabetes (112); however, it is debated as to whether or not colonized patients are predisposed to increased complications (113) such as bullous erysipelas, carbuncles, or perifollicular abscesses. Regardless, it is important that appropriate precautions are taken in these patients and that antibiotics are selected that account for antimicrobial resistance.

 

Infection of the foot is the most common type of soft tissue infection in patients with diabetes. If not managed properly, diabetic foot infections can become severe, possibly leading to sepsis, amputation, or even death. Although less severe, the areas between the toes and the toenails are also frequently infected in patients with diabetes. Infections can stem from monomicrobial or polymicrobial etiologies. Staphylococcal infections are the most common (114), although complications with infection by Pseudomonas aeruginosa are also common (115). Pseudomonal infection of the toenail may present with a green discoloration, which may become more pronounced with the use of a Wood’s light. Treatment frequently requires coordination of care from multiple medical providers. Topical or oral antibiotics and surgical debridement may be indicated depending on the severity of the infection.

 

Necrotizing fasciitis is an acute life-threatening infection of the skin and the underlying tissue. Those with poorly controlled diabetes are at an increased risk for necrotizing fasciitis. Necrotizing fasciitis presents early with erythema, induration, and tenderness which may then progress within days to hemorrhagic bullous. Patients will classically present with severe pain out of proportion to their presentation on physical exam. Palpation of the affected area often illicit crepitus. Involvement can occur on any part of the body but normally occurs in a single area, most commonly affecting the lower extremities. Fournier’s gangrene refers to necrotizing fasciitis of the perineum or genitals, often involving the scrotum and spreading rapidly to adjacent tissues. The infection in patients with diabetes is most often polymicrobial. Complications of necrotizing fasciitis include thrombosis, gangrenous necrosis, sepsis, and organ failure. Necrotizing fasciitis has a mortality rate of around twenty percent (116). In addition, those patients with diabetes and necrotizing fasciitis are more likely to require amputation during their treatment (117). Treatment is emergent and includes extensive surgical debridement and broad-spectrum antibiotics.

 

Erythrasma is a chronic asymptomatic cutaneous infection, most often attributed to Corynebacterium minutissiumum. Diabetes mellitus, as well as obesity and older age are associated with erythrasma. Erythrasma presents with non-pruritic non-tender clearly demarcated red-brown finely scaled patches or plaques. These lesions are commonly located in intriginuous areas such as the axilla or groin. Given the appearance and location, erythrasma can be easily mistaken for tinea or Candidia infection; in such cases, the presence of coral-red fluorescence under a Wood’s light can confirm the diagnosis of erythrasma. Treatment options include topical erythromycin or clindamycin, Whitfield’s ointment, and sodium fusidate ointment. More generalized erythrasma may respond better to oral erythromycin.

 

Malignant otitis externa is a rare but serious infection of the external auditory canal that occurs most often in those with a suppressed immune system, diabetes mellitus, or of older age. Malignant otitis externa develops as a complication of otitis externa and is associated with infection by Pseudomonas aeruginosa. Patients with malignant otitis externa present with severe otalgia and purulent otorrhea. The infection can spread to nearby structures and cause complications such as chondritis, osteomyelitis, meningitis, or cerebritis. If untreated, malignant otitis externa has a mortality rate of about 50%; however, with aggressive treatment the mortality rate can been reduced to 10% to 20% (118). Treatment involves long-term systemic antibiotics with appropriate pseudomonal coverage, hyperbaric oxygen, and possibly surgical debridement.

 

FUNGAL

 

Candidiasis is a frequent presentation in patients with diabetes. Moreover, asymptomatic patients presenting with recurrent candidiasis should be evaluated for diabetes mellitus. Elevated salivary glucose concentrations (119) and elevated skin surface pH in the intertriginous regions of patients with diabetes (120) may promote an environment in which candida can thrive. Candida infection can involve the mucosa (e.g., thrush, vulvovaginitis), intertriginous areas (e.g., intertrigo, erosion interdigital, balanitis), or nails (e.g., paronychia). Mucosal involvement presents with pruritus, erythema, and white plaques which can be removed when scraped. Intertriginous Candida infections may be pruritic or painful and present with red macerated, fissured plaques with satellite vesciulopustules. Involvement of the nails may present with periungual inflammation or superficial white spots. Onchyomycosis may be due to dermatophytes (discussed below) or Candidal infection. Onchomycosis, characterized by subungual hyperkeratosis and oncholysis, is present in nearly one in two patients with type 2 diabetes mellitus. Candidiasis is treated with topical or oral antifungal agents. Patients also benefit from improved glycemic control and by keeping the affected areas dry.

 

Although it remains controversial, dermatophyte infections appear to be more prevalent among patients with diabetes (121-123). Various regions of the body may be affected but tinea pedis (foot) is the most common dermatophyte infection effecting patients; it presents with pruritus or pain and erythematous keratotic or bullous lesions. Relatively benign dermatophyte infections like tinea pedis can lead to serious sequela, such as secondary bacterial infection, fungemia, or sepsis, in patients with diabetes if not treated hastily. Patients with diabetic neuropathy may be especially vulnerable (124). Treatment may include topical or systemic antifungal medications depending on the severity.

 

Mucormycosis is a serious infection that is associated with type 1 diabetes mellitus, particularly common in those who develop diabetic ketoacidosis. A variety of factors including hyperglycemia and a lower pH, create an environment in which Rhizopus oryzae, a common pathogen responsible for mucormycosis, can prosper. Mucormycosis may present in different ways. Rhino-orbital-cerebral mucormycosis is the most common presentation; it develops quickly and presents with acute sinusitis, headache, facial edema, and tissue necrosis. The infection may worsen to cause extensive necrosis and thrombosis of nearby structures such as the eye. Mucormycosis should be treated urgently with surgical debridement and intravenous amphotericin B. When it is not suitable to administer amphotericin B in patients, the alternative use of new triazoles, posaconazole and isavuconazole, may be beneficial treatments (124A).

 

Lastly, abnormal toe web findings (e.g., maceration, scale, or erythema) may be an early marker of irregularities in glucose metabolism and of undiagnosed diabetes mellitus (125). Additionally, such findings may be a sign of epidermal barrier disruption, a precursor of infection (125).

 

CUTANEOUS CHANGES ASSOCIATED WITH DIABETES MEDICATIONS

 

Insulin

 

A number of localized changes are associated with the subcutaneous injection of insulin. The most common local adverse effect is lipohypertrophy, which affects less than thirty percent of patients with diabetes that use insulin (126,127). Lipohypertrophy is characterized by localized adipocyte hypertrophy and presents with soft dermal nodules at injection sites. Continued injection of insulin at sites of lipohypertrophy can result in delayed systemic insulin absorption and capricious glycemic control. With avoidance of subcutaneous insulin at affected sites, lipohypertrophy normally improves over the course of a few months.

 

Furthermore, lipoatrophy is an uncommon cutaneous finding which occurred more frequently prior to the introduction of modern purified forms of insulin. Lipoatrophy presents at insulin injection sites over a period of months with round concave areas of adipose tissue atrophy. Allergic reactions to the injection of insulin may be immediate (within one hour) or delayed (within one day) and can present with localized or systemic symptoms. These reactions may be due to a type one hypersensitivity reaction to insulin or certain additives. However, allergic reactions to subcutaneous insulin are rare, with systemic allergic reactions occurring in only 0.01% of patients (126). Other cutaneous changes at areas of injection include the development of pruritus, induration, erythema, nodular amyloidosis, or calcification.

 

Oral Medications

 

Oral hypoglycemic agents may cause a number of different cutaneous adverse effects such as erythema multiforme or urticaria. DPP-IV inhibitors, such as vildagliptin, can be associated with inflamed blistering skin lesions, including bullous pemphigoid and Stevens-Johnson syndrome, as well as, angioedema (128,129). Allergic skin and photosensitivity reactions may occur with sulfonylureas (130). The sulfonylureas, chlorpropamide and tolbutamide, are associated with the development of a maculopapular rash during the initial two months of treatment; the rash quickly improves with stoppage of the medication (131,132). In certain patients with genetic predispositions, chlorpropamide may also cause acute facial flushing following alcohol consumption (133). SGLT-2 inhibitors have been associated with an increased risk of genital fungal infections and Fournier’s gangrene (134) (for details see Endotext chapter Oral and Injectable (Non-Insulin) Pharmacological Agents for the Treatment of Type 2 Diabetes) (135).

 

CONCLUSION

 

Diabetes mellitus is associated with a broad array of dermatologic conditions (Table 1). Many of the sources describing dermatologic changes associated with diabetes mellitus are antiquated; larger research studies utilizing modern analytic tools are needed to better understand the underlying pathophysiology and treatment efficacy. Although each condition may respond to a variety of specific treatments, many will improve with improved glycemic control. Hence, patient education and lifestyle changes are key in improving the health and quality of life of patients with diabetes mellitus.

 

Table 1. Frequent Skin Manifestations of Diabetes Mellitus

DISEASE

APPEARANCE

COMMON LOCATIONS

SYMPTOMS

TREATMENT

Acanthosis Nigricans

Multiple poorly demarcated plaques with grey to dark-brown hyperpigmentation, and a thickened velvety to verrucous texture

Back of the neck, axilla, elbows, palmer hands, inframammary creases, umbilicus, groin

Typically, asymptomatic

Improved glycemic control, oral retinoids, ammonium lactate, retinoic acid, salicylic acid

Diabetic Dermopathy

Rounded, dull, red papules that progressively evolve over one-to-two weeks into well-circumscribed, atrophic, brown macules with a fine scale; lesions present in different stages of evolution at the same time

Pretibial area, lateral meoli, thighs

Typically, asymptomatic

Self-resolving

Diabetic Foot Syndrome

Chronic ulcers, secondary infection, diabetic neuro-osteoarthropathy, clawing deformity

Feet

Typically, asymptomatic but may have abnormal gait

Interdisciplinary team-based approach involving daily surveillance, appropriate foot hygiene, proper footwear/walker, wound care, antibiotics, wound debridement, surgery

Scleroderma-like Skin Changes

Slowly developing painless, indurated, occasionally waxy appearing, thickened skin

Acral areas: dorsum of the fingers, proximal interphalangeal areas, metacarpophalangeal joints

Typically, asymptomatic but may have reduced range of motion

Improved glycemic control, aldose reductase inhibitors, physical therapy

Ichthyosiform Skin Changes

Large bilateral areas of dryness and scaling (may be described as “fish scale” skin)

Anterior shins, hands, feet

Typically, asymptomatic

Emollients, Keratolytics

Xerosis

Abnormally dry skin that may also present with scaling or fissures

Most common on the feet

Typically, asymptomatic

Emollients

Pruritus

Normal or excoriated skin

Often localized to the scalp, ankles, feet, trunk, or genitalia; however, it may be generalized

Pruritus

Topical capsaicin, topical ketamine-amitriptyline-lidocaine, oral anticonvulsants, antifungals

 

REFERENCES

 

  1. Meurer M, Stumvoll M, Szeimies R-M. Hautveränderungen bei Diabetes mellitus. Der Hautarzt 2004;55(5):428-35
  2. Stuart CA, Gilkison CR, Smith MM, Bosma AM, Keenan BS, Nagamani M. Acanthosis nigricans as a risk factor for non-insulin dependent diabetes mellitus. Clinical Pediatrics 1998;37(2):73-79
  3. Stuart CA, Driscoll MS, Lundquist KF, Gilkison CR, Shaheb S, Smith MM. Acanthosis nigricans. Journal of basic and clinical physiology and pharmacology 1998;9(2-4):407-18
  4. Yeh JS, Munn SE, Plunkett TA, Harper PG, Hopster DJ, du Vivier AW. Coexistence of acanthosis nigricans and the sign of Leser-Trelat in a patient with gastric adenocarcinoma: a case report and literature review. Journal of the American Academy of Dermatology 2000;42(2):357-62
  5. Bhagyanathan M, Dhayanithy D, Parambath VA, Bijayraj R. Acanthosis nigricans: A screening test for insulin resistance–An important risk factor for diabetes mellitus type-2. Journal of family medicine and primary care 2017;6(1):43
  6. Hermanns-Lê T, Scheen A, Piérard GE. Acanthosis nigricans associated with insulin resistance. American journal of clinical dermatology 2004;5(3):199-203
  7. Higgins SP, Freemark M, Prose NS. Acanthosis nigricans: a practical approach to evaluation and management. Dermatology online journal 2008;14(9)
  8. Blobstein SH. Topical therapy with tretinoin and ammonium lactate for acanthosis nigricans associated with obesity. Cutis 2003;71(1):33-34
  9. Ehsani A, Noormohammadpour P, Goodarzi A, et al. Comparison of long-pulsed alexandrite laser and topical tretinoin-ammonium lactate in axillary acanthosis nigricans: A case series of patients in a before-after trial. Caspian journal of internal medicine 2016;7(4):290
  10. Sinha S, Schwartz RA. Juvenile acanthosis nigricans. Journal of the American Academy of Dermatology 2007;57(3):502-08
  11. Bustan RS, Wasim D, Yderstræde KB, Bygum A. Specific skin signs as a cutaneous marker of diabetes mellitus and the prediabetic state-a systematic review. Danish medical journal 2017;64(1)
  12. Morgan AJ, Schwartz RA. Diabetic dermopathy: A subtle sign with grave implications. Journal of the American Academy of Dermatology 2008;58(3):447-51
  13. Romano G, Moretti G, Di Benedetto A, et al. Skin lesions in diabetes mellitus: prevalence and clinical correlations. Diabetes research and clinical practice 1998;39(2):101-06
  14. Huntley AC. The cutaneous manifestations of diabetes mellitus. Journal of the American Academy of Dermatology 1982;7(4):427-55
  15. Houck GM, Morgan MB. A reappraisal of the histologic findings of pigmented pretibial patches of diabetes mellitus. Journal of cutaneous pathology 2004;31(2):141-44
  16. McCASH S, Emanuel PO. Defining diabetic dermopathy. The Journal of dermatology 2011;38(10):988-92
  17. Kaňková K, Záhejský J, Márová I, et al. Polymorphisms in the RAGE gene influence susceptibility to diabetes-associated microvascular dermatoses in NIDDM. Journal of Diabetes and its Complications 2001;15(4):185-92
  18. Kiziltan M, Benbir G. Clinical and nerve conduction studies in female patients with diabetic dermopathy. Acta diabetologica 2008;45(2):97-105
  19. Amin N, Doupis J. Diabetic foot disease: from the evaluation of the “foot at risk” to the novel diabetic ulcer treatment modalities. World journal of diabetes 2016;7(7):153
  20. Sämann A, Tajiyeva O, Müller N, et al. Prevalence of the diabetic foot syndrome at the primary care level in Germany: a cross‐sectional study. Diabetic Medicine 2008;25(5):557-63
  21. Singh N, Armstrong DG, Lipsky BA. Preventing foot ulcers in patients with diabetes. Jama 2005;293(2):217-28
  22. Boulton AJ, Vileikyte L, Ragnarson-Tennvall G, Apelqvist J. The global burden of diabetic foot disease. The Lancet 2005;366(9498):1719-24
  23. Mlinarić-Missoni E, Kalenić S, Važić-Babić V. Species distribution and frequency of isolation of yeasts and dermatophytes from toe webs of diabetic patients. Acta dermatovenerologica Croatica 2005;13(2):0-0
  24. Davis MD, O'fallon WM, Rogers III RS, Rooke TW. Natural history of erythromelalgia: presentation and outcome in 168 patients. Archives of Dermatology 2000;136(3):330-36
  25. Jeffcoate WJ, Harding KG. Diabetic foot ulcers. The lancet 2003;361(9368):1545-51
  26. Lobmann R, Schultz G, Lehnert H. Proteases and the diabetic foot syndrome: mechanisms and therapeutic implications. Diabetes care 2005;28(2):461-71
  27. Volmer-Thole M, Lobmann R. Neuropathy and diabetic foot syndrome. International journal of molecular sciences 2016;17(6):917
  28. Sandireddy R, Yerra VG, Areti A, Komirishetty P, Kumar A. Neuroinflammation and oxidative stress in diabetic neuropathy: futuristic strategies based on these targets. International journal of endocrinology 2014;2014

28A. Theocharidis G, Thomas BE, Sarkar D, Mumme HL, Pilcher WJR, Dwivedi B, Sandoval-Schaefer T, Sîrbulescu RF, Kafanas A, Mezghani I, Wang P, Lobao A, Vlachos IS, Dash B, Hsia HC, Horsley V, Bhasin SS, Veves A, Bhasin M. Single cell transcriptomic landscape of diabetic foot ulcers. Nat Commun. 2022 Jan 10;13(1):181. doi: 10.1038/s41467-021-27801-8. PMID: 35013299; PMCID: PMC8748704.

  1. Vinik AI, Erbas T. Recognizing and treating diabetic autonomic neuropathy. Cleve Clin J Med 2001;68(11):928-30, 32, 34-44
  2. Peripheral Arterial Disease in People With Diabetes. Diabetes Care 2003;26(12):3333-41 .

30A. Lanting SM, Chan TL, Casey SL, Peterson BJ, Chuter VH. Cutaneous microvascular reactivity in Charcot neuroarthropathy: a systematic review and meta-analysis. J Foot Ankle Res. 2022 Mar 1;15(1):17. doi: 10.1186/s13047-022-00522-x. PMID: 35232466; PMCID: PMC8886937.

  1. Khanna S, Biswas S, Shang Y, et al. Macrophage dysfunction impairs resolution of inflammation in the wounds of diabetic mice. PloS one 2010;5(3):e9539
  2. Ahn C, Yosipovitch G, Huang W. Diabetes and the Skin. In: Callen J, Jorizzo J, Zone J, Piette W, Rosenbach M, Vleugels RA, eds. Dermatological Signs of Systemic Disease. Edinburgh: Elsevier, 2017:205-14.
  3. Yosipovitch G, Hodak E, Vardi P, et al. The prevalence of cutaneous manifestations in IDDM patients and their association with diabetes risk factors and microvascular complications. Diabetes care 1998;21(4):506-09
  4. Brik R, Berant M, Vardi P. The scleroderma‐like syndrome of insulin‐dependent diabetes mellitus. Diabetes/Metabolism Research and Reviews 1991;7(2):121-28
  5. Yosipovitch G, Yosipovitch Z, Karp M, Mukamel M. Trigger finger in young patients with insulin dependent diabetes. The Journal of rheumatology 1990;17(7):951-52
  6. Kim RP, Edelman SV, Kim DD. Musculoskeletal complications of diabetes mellitus. Clinical Diabetes 2001;19(3):132-35
  7. Haustein UF. Scleroderma‐like lesions in insulin‐dependent diabetes mellitus. Journal of the European Academy of Dermatology and Venereology 1999;13(1):50-53
  8. Yosipovitch G, Loh KC, Hock OB. Medical pearl: Scleroderma-like skin changes in patients with diabetes mellitus. Journal of the American Academy of Dermatology 2003;49(1):109-11
  9. Rho YW, Suhr KB, Lee JH, Park JK. A clinical observation of scleredema adultorum and its relationship to diabetes. The Journal of dermatology 1998;25(2):103-07
  10. Sawatkar G, Kanwar A, Dogra S, Bhadada S, Dayal D. Spectrum of cutaneous manifestations of type 1 diabetes mellitus in 500 south Asian patients. British Journal of Dermatology 2014;171(6):1402-06
  11. Gerrits EG, Landman GW, Nijenhuis-Rosien L, Bilo HJ. Limited joint mobility syndrome in diabetes mellitus: A minireview. World journal of diabetes 2015;6(9):1108
  12. Fitzgibbons PG, Weiss A-PC. Hand manifestations of diabetes mellitus. The Journal of hand surgery 2008;33(5):771-75
  13. Abate M, Schiavone C, Pelotti P, Salini V. Limited joint mobility in diabetes and ageing: recent advances in pathogenesis and therapy: SAGE Publications Sage UK: London, England, 2010.
  14. Frost D, Beischer W. Limited joint mobility in type 1 diabetic patients. Diabetes care 2001;24(1):95-99
  15. Lopez-Martin I, Benito Ortiz L, Rodriguez-Borlado B, Cano Langreo M, Garcia-Martinez FJ, Martin Rodriguez MF. (Association between limited joint mobility syndrome and risk of accidental falls in diabetic patients). Semergen 2015;41(2):70-5
  16. Lindsay JR, Kennedy L, Atkinson AB, et al. Reduced prevalence of limited joint mobility in type 1 diabetes in a UK clinic population over a 20-year period. Diabetes Care 2005;28(3):658-61
  17. Rongioletti F, Kaiser F, Cinotti E, et al. Scleredema. A multicentre study of characteristics, comorbidities, course and therapy in 44 patients. Journal of the European Academy of Dermatology and Venereology 2015;29(12):2399-404
  18. Cole GW, Headley J, Skowsky R. Scleredema Diabeticorum: A Common and Distinct Cutaneous Manifestation of Diabetes Mellitus. Diabetes Care 1983;6(2):189-92.
  19. Carrington PR, Sanusi I, Winder PR, Turk LL, Jones C, Millikan LE. Scleredema adultorum. International journal of dermatology 1984;23(8):514-22
  20. Martin C, Requena L, Manrique K, Manzarbeitia F, Rovira A. Scleredema diabeticorum in a patient with type 2 diabetes mellitus. Case reports in endocrinology 2011;2011
  21. Varga J, Gotta S, Li L, Sollberg S, Leonardo M. Scleredema adultorum: case report and demonstration of abnormal expression of extracellular matrix genes in skin fibroblasts in vivo and in vitro. British Journal of Dermatology 1995;132(6):992-99
  22. Ferreli C, Gasparini G, Parodi A, Cozzani E, Rongioletti F, Atzori L. Cutaneous Manifestations of Scleroderma and Scleroderma-Like Disorders: a Comprehensive Review. Clinical Reviews in Allergy & Immunology 2017:1-31
  23. Bray SM, Varghese S, English JC. Ultrasonic massage and physical therapy for scleredema: improving activities of daily living. Archives of dermatology 2010;146(4):453-54
  24. Reid SD, Ladizinski B, Lee K, Baibergenova A, Alavi A. Update on necrobiosis lipoidica: a review of etiology, diagnosis, and treatment options. Journal of the American Academy of Dermatology 2013;69(5):783-91
  25. O’toole E, Kennedy U, Nolan J, Young M, Rogers S, Barnes L. Necrobiosis lipoidica: only a minority of patients have diabetes mellitus. Br J Dermatol 1999;140(2):283-6
  26. Muller SA, Winkelmann R. Necrobiosis lipoidica diabeticorum: a clinical and pathological investigation of 171 cases. Archives of dermatology 1966;93(3):272-81

56A. Severson KJ, Patel MH, Brumfiel CM, Breen I, Butterfield RJ, Nelson SA, Sekulic A, Pittelkow MR, Mangold AR. Comorbidities and diabetic complications in patients with necrobiosis lipoidica. J Am Acad Dermatol. 2022 Apr;86(4):891-894. doi: 10.1016/j.jaad.2021.03.026. Epub 2021 Mar 17. PMID: 33722550.

  1. Kota SK, Jammula S, Kota SK, Meher LK, Modi KD. Necrobiosis lipoidica diabeticorum: A case-based review of literature. Indian journal of endocrinology and metabolism 2012;16(4):614
  2. Lopez PR, Leicht S, Sigmon JR, Stigall L. Bullosis diabeticorum associated with a prediabetic state. Southern medical journal 2009;102(6):643-44
  3. Larsen K, Jensen T, Karlsmark T, Holstein PE. Incidence of bullosis diabeticorum–a controversial cause of chronic foot ulceration. International wound journal 2008;5(4):591-96
  4. Lipsky BA, Baker PD, Ahroni JH. Diabetic bullae: 12 cases of a purportedly rare cutaneous disorder. International journal of dermatology 2000;39(3):196-200
  5. Patel N, Spencer LA, English JC, Zirwas MJ. Acquired ichthyosis. Journal of the American Academy of Dermatology 2006;55(4):647-56
  6. Pavlović MD, Milenković T, Dinić M, et al. The prevalence of cutaneous manifestations in young patients with type 1 diabetes. Diabetes care 2007;30(8):1964-67
  7. Goyal A, Raina S, Kaushal SS, Mahajan V, Sharma NL. Pattern of cutaneous manifestations in diabetes mellitus. Indian journal of dermatology 2010;55(1):39

63A. Iacopi E, Riitano N, Dini V, Berta R, Pieruzzi L, Janowska A, Anselmino M, Piaggesi A, Romanelli M. Using Skin Bioengineering to Highlight How Weight and Diabetes Mellitus Modify the Skin in the Lower Limbs of Super-Obese Patients. Diabetes Metab Syndr Obes. 2020 Mar 16;13:729-738. doi: 10.2147/DMSO.S229042. PMID: 32214836; PMCID: PMC7083633.

  1. Pavicic T, Korting HC. Xerosis and callus formation as a key to the diabetic foot syndrome: dermatologic view of the problem and its management. JDDG: Journal der Deutschen Dermatologischen Gesellschaft 2006;4(11):935-41
  2. Saray Y, Seçkin D, Bilezikçi B. Acquired perforating dermatosis: clinicopathological features in twenty‐two cases. Journal of the European Academy of Dermatology and Venereology 2006;20(6):679-88
  3. Karpouzis A, Giatromanolaki A, Sivridis E, Kouskoukis C. Acquired reactive perforating collagenosis: current status. The Journal of dermatology 2010;37(7):585-92
  4. Poliak SC, Lebwohl MG, Parris A, Prioleau PG. Reactive perforating collagenosis associated with diabetes mellitus. New England Journal of Medicine 1982;306(2):81-84
  5. Abdelbaqi‐Salhab M, Shalhub S, Morgan MB. A current review of the cutaneous manifestations of renal disease. Journal of cutaneous pathology 2003;30(9):527-38
  6. Robles-Mendez J, Vazquez-Martinez O, Ocampo-Candiani J. Skin manifestations of chronic kidney disease. Actas Dermo-Sifiliográficas 2015;106(8):609-22
  7. Morton C, Henderson I, Jones M, Lowe J. Acquired perforating dermatosis in a British dialysis population. British Journal of Dermatology 1996;135(5):671-77

70A. Forouzandeh M, Stratman S, Yosipovitch G. The treatment of Kyrle's disease: a systematic review. J Eur Acad Dermatol Venereol. 2020 Jul;34(7):1457-1463. doi: 10.1111/jdv.16182. Epub 2020 Feb 5. PMID: 31919924.

  1. Van Hattem S, Bootsma AH, Thio HB. Skin manifestations of diabetes. Cleve Clin J Med 2008;75(11):772-74
  2. Wagner G, Sachse MM. Acquired reactive perforating dermatosis. JDDG: Journal der Deutschen Dermatologischen Gesellschaft 2013;11(8):723-29 doi: 10.1111/ddg.12131(published Online First: Epub Date)|.

72A. Ying Y, Shuang C, Zhen-Ying Z. Dupilumab may be an alternative option in the treatment of acquired reactive perforating collagenosis combined with AD. Immun Inflamm Dis. 2022 Mar;10(3):e574. doi: 10.1002/iid3.574. Epub 2021 Dec 24. PMID: 34953055; PMCID: PMC8926492.

  1. Sugandhan S, Khandpur S, Sharma VK. Familial chylomicronemia syndrome. Pediatric dermatology 2007;24(3):323-25
  2. Mahajan S, Koranne R, Sharma S. Cutaneous manifestation of diabetes melitus. Indian Journal of Dermatology, Venereology, and Leprology 2003;69(2):105
  3. Ahmed K, Muhammad Z, Qayum I. Prevalence of cutaneous manifestations of diabetes mellitus. J Ayub Med Coll Abbottabad 2009;21(2):76-9
  4. Vergès B. Pathophysiology of diabetic dyslipidaemia: where are we? Diabetologia 2015;58(5):886-99
  5. Naik NS. Eruptive xanthomas. Dermatology online journal 2001;7(2)
  6. Zaremba J, Zaczkiewicz A, Placek W. Eruptive xanthomas. Advances in Dermatology and Allergology/Postȩpy Dermatologii i Alergologii 2013;30(6):399
  7. Parker F. Xanthomas and hyperlipidemias. Journal of the American Academy of Dermatology 1985;13(1):1-30
  8. Ales Z, Zeman M, Slaby A, Vecka M. Xanthomas: clinical and pathophysiological relations. Biomedical Papers 2014;158(2):181-88
  9. Margolis J. Letter: Skin tags in diabetes mellitus. N Engl J Med 1976;295:172-3
  10. Levy L, Zeichner JA. Dermatologic manifestation of diabetes. Journal of diabetes 2012;4(1):68-76
  11. Yamaoka H, Sasaki H, Yamasaki H, et al. Truncal pruritus of unknown origin may be a symptom of diabetic polyneuropathy. Diabetes care 2010;33(1):150-55
  12. Valdes-Rodriguez R, Mollanazar NK, González-Muro J, et al. Itch prevalence and characteristics in a Hispanic geriatric population: a comprehensive study using a standardized itch questionnaire. Acta dermato-venereologica 2015;95(4):417-21

84A. Yang QP, Chen YY, Li Z, Xu M. Major Risk Factors Analysis of Pruritus Complicated by Type 2   Diabetes Mellitus and the Effect of Comprehensive Nursing Intervention. Front Surg. 2022 Feb 15;9:842884. doi: 10.3389/fsurg.2022.842884. PMID: 35242808; PMCID: PMC8887712.

  1. Guarneri C, Guarneri F, Borgia F, Vaccaro M. Finger pebbles in a diabetic patient: Huntley's papules. International journal of dermatology 2005;44(9):755-56
  2. Tabor CA, Parlette EC. Cutaneous manifestations of diabetes: signs of poor glycemic control or new-onset disease. Postgraduate medicine 2006;119(3):38-44
  3. Murphy-Chutorian B, Han G, Cohen SR. Dermatologic manifestations of diabetes mellitus. Endocrinology and Metabolism Clinics 2013;42(4):869-98
  4. Liau M, Long V, Yang S, Tan K, Aw D. Pigmented purpuric dermatosis in Singapore: a clinic-epidemiological characterisation. Hong Kong Journal of Dermatology & Venereology 2016;24(2):65-69
  5. Lithner F. Purpura, pigmentation and yellow nails of the lower extremities in diabetics. Journal of Internal Medicine 1976;199(1‐6):203-08
  6. Demirseren DD, Emre S, Akoglu G, et al. Relationship between skin diseases and extracutaneous complications of diabetes mellitus: clinical analysis of 750 patients. American journal of clinical dermatology 2014;15(1):65-70
  7. Serrao R, Zirwas M, English JC. Palmar erythema. American journal of clinical dermatology 2007;8(6):347-56
  8. Landau J, Davis E. The small blood-vessels of the conjunctiva and nailbed in diabetes mellitus. The Lancet 1960;276(7153):731-34
  9. Oumeish OY. Skin disorders in patients with diabetes. Clinics in dermatology 2008;26(3):235-42
  10. Duff M, Demidova O, Blackburn S, Shubrook J. Cutaneous manifestations of diabetes mellitus. Clinical Diabetes 2015;33(1):40-48
  11. Bristow I. Non‐ulcerative skin pathologies of the diabetic foot. Diabetes/metabolism research and reviews 2008;24(S1)

95A. Yesudian PD, Nwabudike LC, de Berker D. Nail changes in diabetes. Clin Exp Dermatol. 2022 Jan;47(1):9-15. doi: 10.1111/ced.14859. Epub 2021 Sep 21. PMID: 34293827.

  1. Mendes AL, Miot HA, Haddad Junior V. Diabetes mellitus and the skin. Anais brasileiros de dermatologia 2017;92(1):8-20
  2. Yun JH, Lee JY, Kim MK, et al. Clinical and pathological features of generalized granuloma annulare with their correlation: a retrospective multicenter study in Korea. Annals of dermatology 2009;21(2):113-19
  3. Cyr PR. Diagnosis and management of granuloma annulare. American family physician 2006;74(10)
  4. Armstrong AW, Harskamp CT, Armstrong EJ. Psoriasis and the risk of diabetes mellitus: A systematic review and meta-analysis. JAMA Dermatology 2013;149(1):84-91
  5. Seyhan M, Özcan H, Sahin I, Bayram N, Karincaoğlu Y. High prevalence of glucose metabolism disturbance in patients with lichen planus. Diabetes research and clinical practice 2007;77(2):198-202
  6. Guggenheimer J, Moore PA, Rossie K, et al. Insulin-dependent diabetes mellitus and oral soft tissue pathologies: I. Prevalence and characteristics of non-candidal lesions. Oral Surgery, Oral Medicine, Oral Pathology, Oral Radiology, and Endodontology 2000;89(5):563-69
  7. Van den Driessche A, Eenkhoorn V, Van Gaal L, De Block C. Type 1 diabetes and autoimmune polyglandular syndrome: a clinical review. Neth J Med 2009;67(11):376-87
  8. Von der Werth J, Williams H. The natural history of hidradenitis suppurativa. Journal of the European Academy of Dermatology and Venereology 2000;14(5):389-92
  9. Bui T-L, Silva-Hirschberg C, Torres J, Armstrong AW. Hidradenitis suppurativa and diabetes mellitus: A systematic review and meta-analysis. Journal of the American Academy of Dermatology 2017
  10. Alikhan A, Lynch PJ, Eisen DB. Hidradenitis suppurativa: a comprehensive review. Journal of the American Academy of Dermatology 2009;60(4):539-61
  11. Eldor R, Glaser B, Fraenkel M, Doviner V, Salmon A, Gross DJ. Glucagonoma and the glucagonoma syndrome–cumulative experience with an elusive endocrine tumour. Clinical endocrinology 2011;74(5):593-98
  12. Wermers RA, Fatourechi V, Wynne AG, Kvols LK, Lloyd RV. The Glucagonoma Syndrome Clinical and Pathologic Features in 21 Patients. Medicine 1996;75(2):53-63
  13. Kovács RK, Korom I, Dobozy A, Farkas G, Ormos J, Kemény L. Necrolytic migratory erythema. Journal of cutaneous pathology 2006;33(3):242-45
  14. Kindmark H, Sundin A, Granberg D, et al. Endocrine pancreatic tumors with glucagon hypersecretion: a retrospective study of 23 cases during 20 years. Medical Oncology 2007;24(3):330-37
  15. Halvorson SA, Gilbert E, Hopkins RS, et al. Putting the pieces together: necrolytic migratory erythema and the glucagonoma syndrome. Journal of general internal medicine 2013;28(11):1525-29
  16. Macedo GMC, Nunes S, Barreto T. Skin disorders in diabetes mellitus: an epidemiology and physiopathology review. Diabetology & metabolic syndrome 2016;8(1):63
  17. Kutlu SS, Cevahir N, Akalin S, et al. Prevalence and risk factors for methicillin-resistant Staphylococcus aureus colonization in a diabetic outpatient population: a prospective cohort study. American journal of infection control 2012;40(4):365-68
  18. Hartemann‐Heurtier A, Robert J, Jacqueminet S, et al. Diabetic foot ulcer and multidrug‐resistant organisms: risk factors and impact. Diabetic Medicine 2004;21(7):710-15
  19. Casqueiro J, Casqueiro J, Alves C. Infections in patients with diabetes mellitus: A review of pathogenesis. Indian journal of endocrinology and metabolism 2012;16(Suppl1):S27
  20. Driscoll JA, Brody SL, Kollef MH. The epidemiology, pathogenesis and treatment of Pseudomonas aeruginosa infections. Drugs 2007;67(3):351-68
  21. Bonne SL, Kadri SS. Evaluation and management of necrotizing soft tissue infections. Infectious Disease Clinics 2017;31(3):497-511
  22. Dworkin M, Westercamp M, Park L, McIntyre A. The epidemiology of necrotizing fasciitis including factors associated with death and amputation. Epidemiology & Infection 2009;137(11):1609-14
  23. Bhandary S, Karki P, Sinha B. Malignant otitis externa: a review. Pacific health dialog 2002;9(1):64-67
  24. Sashikumar R, Kannan R. Salivary glucose levels and oral candidal carriage in type II diabetics. Oral Surgery, Oral Medicine, Oral Pathology, Oral Radiology, and Endodontology 2010;109(5):706-11
  25. Yosipovitch G, Tur E, Cohen O, Rusecki Y. Skin surface pH in intertriginous areas in NIDDM patients: possible correlation to candidal intertrigo. Diabetes Care 1993;16(4):560-63
  26. Saunte DML, Holgersen JB, Hædersdal M, et al. Prevalence of toe nail onychomycosis in diabetic patients. Acta dermato-venereologica 2006;86(5):425-28
  27. Alteras I, Saryt E. Prevalence of pathogenic fungi in the toe-webs and toe-nails of diabetic patients. Mycopathologia 1979;67(3):157-59
  28. Romano C, Massai L, Asta F, Signorini A. Prevalence of dermatophytic skin and nail infections in diabetic patients. Mycoses 2001;44(3‐4):83-86
  29. Rich P. Onychomycosis and tinea pedis in patients with diabetes. Journal of the American Academy of Dermatology 2000;43(5):S130-S34

124A. Sharma A, Goel A. Mucormycosis: risk factors, diagnosis, treatments, and challenges during COVID-19 pandemic. Folia Microbiol (Praha). 2022 Feb 26:1–25. doi: 10.1007/s12223-021-00934-5. Epub ahead of print. PMID: 35220559; PMCID: PMC8881997.

  1. Sinikumpu S-P, Auvinen J, Jokelainen J, et al. Abnormal skin in toe webs is a marker for abnormal glucose metabolism. A cross-sectional survey among 1,849 adults in Finland. Scientific reports 2017;7:9125
  2. Richardson T, Kerr D. Skin-related complications of insulin therapy. American journal of clinical dermatology 2003;4(10):661-67
  3. Lima AL, Illing T, Schliemann S, Elsner P. Cutaneous Manifestations of Diabetes Mellitus: A Review. American Journal of Clinical Dermatology 2017:1-13
  4. Attaway A, Mersfelder TL, Vaishnav S, Baker JK. Bullous pemphigoid associated with dipeptidyl peptidase IV inhibitors. A case report and review of literature. Journal of dermatological case reports 2014;8(1):24
  5. Byrd JS, Minor DS, Elsayed R, Marshall GD. DPP-4 inhibitors and angioedema: a cause for concern? Annals of Allergy, Asthma & Immunology 2011;106(5):436-38
  6. Hitselberger JF, Fosnaugh RP. Photosensitivity due to chlorpropamide. JAMA 1962;180(1):62-63
  7. Walker G, Kinsell LW. Clinical experience with chlorpropamide. California medicine 1961;94(6):344
  8. Walker G, Slater J, Westlake E, Nabarro J. Clinical experience with tolbutamide. British medical journal 1957;2(5040):323
  9. Leslie R, Pyke D. Chlorpropamide-alcohol flushing: a dominantly inherited trait associated with diabetes. Br med J 1978;2(6151):1519-21
  10. Nyirjesy P, Sobel JD, Fung A, et al. Genital mycotic infections with canagliflozin, a sodium glucose co-transporter 2 inhibitor, in patients with type 2 diabetes mellitus: a pooled analysis of clinical studies. Current medical research and opinion 2014;30(6):1109-19
  11. Feingold KR. Oral and Injectable (Non-Insulin) Pharmacological Agents for the Treatment of Type 2 Diabetes. 2021 Aug 28. In: Feingold KR, Anawalt B, Boyce A, et al editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–.

 

Pathophysiology and Treatment of Pancreatic Neuroendocrine Neoplasms (PNENS): New Developments

ABSTRACT  

 

Pancreatic neuroendocrine neoplasms (PNENs) are a heterogenous group of relatively rare pancreatic malignancies with a unique biology and pathophysiology. Over the last few years, there have been significant improvements in imaging and treatment strategies, which have led to advances in patient’s management and quality of life (QOL). Yet, in practice, there are still a number of unanswered questions. For example, it remains a challenge to choose the optimal treatment sequence from the plethora of options and to properly monitor PNEN patients. Therefore, in this chapter, recent advances in the pathophysiology, diagnosis, monitoring, and management of these neoplasms will be summarized and placed in a historical context.

 

INTRODUCTION  

 

Pancreatic neuroendocrine neoplasms (PNENs) are an uncommon subset of neuroendocrine neoplasms (NENs) originating from endocrine cells (1-3). PNENs represent 1-2% of all pancreatic neoplasms and according to the Surveillance, Epidemiology and End Results (SEER) program, the annual age-adjusted incidence has risen from 0.32/100,000 persons in 2004 to 0.48/100,000 persons in 2021 (2, 4-7). Improvements in and a wider availability of high-quality imaging techniques and a well-established classification system are believed to be major factors in the increasing incidence of PNENs (5, 8, 9).

 

PNENs can be divided into both functional (10-40%) and non-functional (60-90%) neoplasms (2, 6, 7, 10, 11). Functional PNENs (F-PNENs) are characterized by a specific clinical course and symptoms due to excessive hormone production (e.g., insulin, gastrin) (10-12). The most frequent, recognized F-PNENs are listed in Table 1 (1). Less common F-PNENs include somatostatinomas, ACTHomas and PNENs that cause carcinoid syndrome, acromegaly, or hypercalcemia (2). Patients with non-functional PNENs (NF-PNENs) lack symptoms related to clinical hormonal syndromes and are therefore usually diagnosed at a more advanced stage with characteristically large primary tumors (70% >5 cm) and liver metastasis in more than 60% of the cases (2, 9, 12, 13). NF-PNENs are hence frequently discovered by chance on imaging studies performed due to nonspecific abdominal pain, often caused by tumor bulk (2, 9, 12, 14). Although NF-PNENs do not secrete peptides causing clinical syndromes, they characteristically secrete a number of other peptides including chromogranin A (CgA) and pancreatic polypeptide (PP). However, elevated levels of PP or CgA are not specific for NF-PNENs as they are also observed in patients with renal failure and inflammatory conditions (2, 9, 12-14).

 

Table 1. Overview of Recognized Functional PNENs and Their Characteristics

Tumor

[syndrome]

Hormone

Clinical symptoms

Biochemical diagnosis

Insulinoma

[Whipple’s triad]

Insulin

Hypoglycemia

At hypoglycemia:

Insulin > 6 µU/L

Glucose 40 mg/dL

C-peptide 0.6 ng/mL

Proinsulin ³ 20 pmol/L

Gastrinoma

[Zollinger-Ellison]

Gastrin

Abdominal pain, Gastroesophageal reflux, Diarrhea, Duodenal ulcers

Serum fasting gastrin level ³ 10 times normal range

VIPoma

[Verner-Morrison]

Vasoactive intestinal peptide (VIP)

Severe watery diarrhea, Hypokalemia

Fasting serum VIP > 60 pmol/L

Glucagonoma

Glucagon

Rash, Glucose intolerance (diabetes), Necrolytic migratory erythema, Weight loss

Fasting glucagon > 500 pg/mL

Note: This table was assembled based on information from Gastroenterology, Metz D. and Jensen R., Gastrointestinal neuroendocrine tumors: Pancreatic Endocrine tumors, 1469-1492 © 2008 (2) and World Journal of Gastroenterology, Ma Z., Gong Y., Zhuang H. et al., Pancreatic neuroendocrine tumors: A review of serum biomarkers, staging and management, 2305-2322 © 2020 (7) and Current Opinion in Gastroenterology, Perri G., Prakash L. and Katz M., Pancreatic neuroendocrine tumors, 468-477 © 2019 (3).  

 

CLASSIFICATION AND STAGING  

 

The World Health Organization (WHO) classification from 2019 (Table 2) takes into account both differentiation status and proliferation rate of the tumor. The former is determined through a histological examination of tumor morphology in which well-differentiated neuroendocrine tumors (NETs) can be distinguished from poorly differentiated neuroendocrine carcinomas (NECs). A grade is then assigned based on the proliferation rate assessed via Ki-67 index and mitotic count. Well-differentiated NETs can be divided into low grade (G1), intermediate grade (G2), and high grade (G3) tumors that have respective Ki-67 values of <3%, 3-20%, and >20% or mitotic counts of <2, 2-20, and >20 per 2mm³ (10 high power fields (HPF)). In the poorly-differentiated NEC group (small and large cell types), only high grade G3 tumors with a Ki-67 value >20 are found. In addition, neoplasms exist that consist of neuroendocrine cells as well as non-neuroendocrine adenocarcinoma or squamous carcinoma cells (i.e., mixed non-neuroendocrine-neuroendocrine neoplasms (MiNEN)) (3, 6-8, 15, 16). Depending on tumor grade and primary site, the 5-year survival varies between 15-95% and median overall survival (OS) from approximately 12 years for patients with G1 to 10 months in patients with G3 PNENs (3, 17). PNENs most often occur sporadically, but can also occur in patients with various inherited disorders (2, 18). For example, PNENs develop in 80-100% of patients with Multiple Endocrine Neoplasia type 1 (MEN1), in 10-17% of patients with von Hippel-Lindau syndrome (VHL), and occasionally in patients with tuberous sclerosis and neurofibromatosis (3, 18). 

 

Table 2. WHO Classification (2019) of PNENs

Type

Differentiation status

Grade

Proliferation rate

Ki-67 (%)

Mitotic count (2mm²)

NEN

Well-differentiated NETs

G1

< 3

< 2

G2

3 – 20

2 – 20

G3

> 20

> 20

Poorly-differentiated NECs

Small cell (SCNECs)

Large cell (LCNECs)

G3

> 20

> 20

MiNEN

NET or NEC + ADC or SCC

G1-G3

See above

See above

Note: This table was adapted from Histopathology, Nagtegaal I., Odze R., Klimstra D. et al., The 2019 WHO classification of tumours of the digestive system, 182-188. © 2019 (16). NEC- neuroendocrine carcinomas; NET- neuroendocrine tumors; ADC- adenocarcinoma cells; SCC- squamous carcinoma cells

 

PNENs are also classified based on the tumor-node-metastasis (TNM) classification which estimates the prognosis of the tumors based on the anatomy of the tumor (3). Previously there was no generally accepted staging system, so in Europe usually the European Neuroendocrine Tumor Society (ENETS) staging system was applied, while in America the America Joint Committee on Cancer (AJCC)/Union for International Cancer Control (UICC) system was being used (19-21). In the 7th edition of the AJCC/UICC, the same ordering system was employed for PNENs as for pancreatic adenocarcinoma (PAAD), but due to biological differences between both tumor types, this staging system proved to have some limitations (19, 20). Consequently, in the revised 8th edition of the AJCC/UICC, the classification system of ENETS was implemented (21). Two research groups demonstrated that the system employed in this 8thedition was superior to that of the 7th edition as well as the ENETS staging system and should be considered as golden standard (20, 22).

 

INDUCTION OF PNENs

 

PNENs are also often referred to as islet cell tumors since it is presumed that they arise from the islets of Langerhans (3, 23, 24). These islets contain A-, B-, D-, D1-, and D2-cells that respectively secrete glucagon, insulin, somatostatin, pancreatic polypeptide, and vasoactive intestinal polypeptide (VIP) (25). Logically, the F-PNENs most definitely arise from these cells, but the cell of origin in NF-PNENs is still a matter of debate (26, 27). Chan et al. revealed that NF-PNENs with ATRX, DAXX, and MEN1 mutations (A-D-M mutant) had a worse clinical outcome than A-D-M wild-type (WT) tumors. In addition, they were able to demonstrate, through RNA sequencing and DNA methylation analysis, that the A-D-M mutant PNENs had high ARX and low PDX1 expression which is consistent with the expression profile found in a-cells (28). Cejas et al. found that NF-PNENs could be divided into two subgroups with epigenomes and transcriptomes very similar to those of a- and b-cells, respectively (29). These findings were confirmed by Di Domenico and colleagues who were able to demonstrate that the genome-wide DNA methylation profiles of NF-PNENs were very consistent with the methylation profiles of a- and b-cells (24). Based on these findings, it was hypothesized that NF-PNENs evolve primarily but not exclusively from the a-cell lineage and b-cell lineage (27).

Figure 1. Visualization of the pancreatic duct glandular structures (PDGs) (arrowheads) in (A) large and (B) small ducts using scanning electron microscopy (SEM). PDGs can occur as single outpouches or form a complex of sac-like dilatations as illustrated in (C). This figure has been adapted from Gastroenterology, Strobel O., Rosow D. E., Rakhlin E. Y., et al., Pancreatic duct glands are distinct ductal compartments that react to chronic injury and mediate Shh-induced metaplasia, 138 (3): 1166-77 © 2010 (30).

Others in turn suggest that PNENs develop from multipotent pancreatic progenitor (MPP) cells in the ductal and islet regions of the pancreas that would be able to generate new pancreatic islet cells (31, 32). However, it remains unclear whether these cells originate in the islets or whether they migrate from the pancreatic ducts to subsequently transform into endocrine cells (33). This hypothesis is strengthened by the fact that early endocrine progenitors in fact appear to originate from a bipotent ductal endocrine progenitor, which in turn originates from MPP cells (34). However, not a lot is known about where these MPP cells are present. One hypothesis states these could be present in the pancreatic duct glandular structures (PDGs) that can be found as specialized compartments with a gland-like outpouching look (Figure 1) in the ductal epithelium (30, 35). The actual origin and location of the MPP that can evolve into islet cells is not known to date and thus needs to be further investigated for a better understanding of the potential origin of PNENs.

 

MOLECULAR (EPI)GENETICS  

 

Genetic Syndromes

 

Although PNENs typically occur sporadically, approximately 10-20% of them develop in the context of hereditary syndromes. The syndrome most at risk for PNEN development is Multiple Endocrine Neoplasia (MEN1) (60%), an autosomal dominant disease caused by inactivating mutations in the MEN1 gene (10, 36-39). MEN1 is a tumor suppressor gene located on chromosome 11q13 that encodes for the nuclear protein menin which plays an important role in the PI3K/Akt/mTOR pathway, histone modifications, DNA repair mechanisms, and cell cycle control (10, 37, 38, 40, 41). In addition, 5 to 18% of the patients with von Hippel-Lindau (VHL) syndrome develop PNENs. These patients carry a germline mutation in the VHL gene located on the short arm of chromosome 3. The VHL protein can be found in different complexes that mediate ubiquitin-mediated degradation and stimulate angiogenesis (10, 42, 43). Other hereditary syndromes at risk include Tuberous Sclerosis (TS) and neurofibromatosis type 1, caused by mutations in TSC1, TSC2 and NF1, respectively (36-39).

 

Sporadic PNENS  

 

Through next-generation sequencing of PNENs, it became apparent that there are distinct genetic differences, strongly depending on differentiation and functionality of the tumor (36).

For example, genetic analyses of F-PNENs revealed that insulinomas are often characterized by a hotspot mutation (p. T372R) in the Yin Yang 1 (YY1) gene in 30% of the Asian and 8-33% of the Western/Caucasian population (10, 36, 44). This recurrent mutation is located in the DNA binding domain of YY1, hence strongly affecting the DNA binding capacity of this transcriptional activator/repressor (44). In NF-PNENs, on the other hand, somatic mutations were most commonly identified in MEN1 (44.1%) followed by DAXX (25%) and ATRX (17.6%) (41). Atrx interacts with DNA methyltransferases (DNMT) 3A and 3L to form the Atrx-DNMT3A-DNMT3L (ADD) complex. This interaction is crucial for maintenance of histone methylation patterns in newly replicated chromatin, hence indirectly ensures correct gene expression. Moreover, Atrx also interacts directly with Daxx. In doing so, Daxx functions as a kind of chaperone for the deposition of histone variant H3.3 at the level of CpG islands, telomeric and ribosomal repeats and the rest of the genome. Consequently, a loss of ATRX and DAXX results in changes in DNA methylation patterns throughout the genome (45). In addition, mutations in PTEN, TSC2, and PIK3CA have already been reported in respectively 7.8%, 8.8% and 1.4% of PNENs, and they all affect the PI3K/Akt/mTOR pathway (41). Later, Scarpa and colleagues identified mutations in DNA repair genes MUTYH, CHEK2, and BRCA2 as well (39).

 

Distinct genetic differences could also be observed between G3 pancreatic NETs (PNETs) and pancreatic NECs (PNECs). The latter do not carry mutations in the known genes for PNETs (MEN1, DAXX, and ATRX), but instead appear to have mutations in TP53, RB1, KRAS, and CDKN2A/p16 (10, 36, 38). Considering that these mutations tend to result in altered protein expression, IHC might facilitate in distinguishing PNETs from PNECs, which have similar Ki-67 values. Nevertheless, results should always be interpreted with caution (10, 36, 46). 

 

Besides point mutations, copy number alterations (CNAs) have also attracted attention. CNA patterns that were frequently identified included whole or partial loss of chromosomes 1, 2, 3, 6, 8, 10, 11, 15, 16, 21 and 22, while gains have been observed in chromosomes 5, 7, 12,14 and 17 (10). Moreover, PNENs appear to display very specific CNA patterns that allow to distinguish PNENs from the more common PAADs. Boons and colleagues therefore developed a classification model, based on tumor tissue, which demonstrated a sensitivity, specificity and area under the curve (AUC) of 100%, 95% and 100% in the validation cohort (47). Benign insulinomas tend to display lower rates of CNAs (36).        

 

Since genes such as MEN1, DAXX and ATRX are of importance in several epigenetic regulatory processes, it was extremely likely that also epigenetic alterations commonly occur in PNENs. In fact, both in hereditary and sporadic PNENs promotor hypermethylation is observed in tumor suppressor genes, which is associated with silencing of gene expression (10, 48). Chan and colleagues checked whether methylation profiles and expression were different in the A-D-M mutated group versus the A-D-M WT group. They observed that both groups clustered in two separate clusters and even revealed that gene expression of the A-D-M mutated group was respectively high and low in the ARX and PDX1 gene and the latter gene also displayed hypermethylation. This profile appeared to be quite similar to that of acells in the pancreas (28). These results were confirmed by Neiman et al., who observed high methylation levels in the PDX1 promotor region in a cells, while β cells tend to have low methylation in this region (49). Based on this PDX1 gene methylation, Boons and colleagues performed unsupervised hierarchical clustering and could subsequently observe two subpopulations, A and B, which respectively contained the a and β cells. Of note, the majority of the mutated PNENs was found in group A confirming the findings of Chan et al. (28, 50). These results suggest that methylation profiling of the PDX1 gene could potentially help to divide PNENs into distinct clinically relevant groups that have different prognosis and risk of relapse (50). Recently, three subgroups (T1, T2 and T3) of PNENs have been identified, based on their methylation profile. Here, the T1 group consisted of the A-D-M WT tumors, while the T2 subgroup encompassed the A-D-M mutated tumors with recurrent chromosomal losses and methylation in the gene body of the MGMT gene. The last group, T3, displayed mutations in MEN1 and recurrent loss of chromosome 11. Tumors found in the latter group tend to have a better prognosis (51).   

 

DIAGNOSIS AND MONITORING           

 

The gold standard for diagnosing PNENs remains an immunohistochemical examination of the tumor tissue, but imaging and serum markers are also extremely important in the diagnostic process. The clinical presentation often determines the sequence of examinations. For example, patients with F-PNENs will usually undergo a biochemical blood analysis first based on their hormonal symptoms, whereas NF-PNENs are often detected by chance on imaging (25, 52, 53).  

 

Immunohistochemistry

 

To correctly classify PNENs, tumor morphology and proliferation rates (Ki-67 and mitotic index) should be evaluated in tissue biopsies. These are usually obtained from surgical specimens, percutaneous core biopsies, or preoperative biopsies (52, 54, 55). The latter were mainly derived from endoscopic ultrasound (EUS) guided fine-needle aspirations (FNA) which, in recent years, have been increasingly replaced by fine-needle biopsies (FNB) as these enable histological tissue samples to be obtained, hence immunohistochemistry (IHC) to be performed (56, 57). This immunohistochemical examination is most often initiated by confirming the neuroendocrine differentiation by checking CgA and synaptophysin (SYP) expression (52, 54). Other markers such as neuron-specific enolase (NSE) and CD56 are less specific, hence less useful (58). Next, tumor morphology is assessed to determine whether the PNEN is well- or poorly-differentiated (Figure 2). In general, well-differentiated PNENs are characterized by uniform cells with a finely granular cytoplasm and round to oval nucleus which are arranged in a trabecular, glandular, or tubuloacinar pattern (54, 59). Moreover, typically all cells have a heterogeneous expression of CgA in their cytoplasm, whereas SYP stains more diffusely. Poorly-differentiated PNECs, on the other hand, consist of atypical neoplastic cells that often lack CgA and even SYP (59, 60). Ultimately, tumors are graded by proliferation rate that is influenced by two parameters, Ki-67 and mitotic count. The latter is usually reported as the number of mitoses per mm² which in practice is often complicated by a limited tissue area. The mitotically active regions are then measured again via IHC to determine the Ki-67 index (Figure 3). It is therefore logical that the Ki-67 index is usually higher than the mitotic count since it considers the entire mitotic process and not just the number of mitoses. If both values assign a different grade to the same tumor, the highest grade, associated with the worst prognosis, is assumed (17, 23, 52, 54).

Figure 2. Hematoxylin-eosin IHC staining of (A) well-differentiated PNET and (B) poorly-differentiated PNEC. This figure has been adapted from Archives of Pathology & Laboratory Medicine, Fang J. M. and Shi J., A clinicopathologic and molecular update of pancreatic neuroendocrine neoplasms with a focus on the new world health organization classification, 143 (11): 1317-1326. © 2019 (59).

Figure 3. (A) PNEN G1 with Ki-67 index of less than 3%. (B) PNEN G2 with Ki-67 index of 3% to 20%. (C) PNEN G3 with Ki-67 index higher than 20%. This figure has been adapted from Archives of Pathology & Laboratory Medicine, Fang J. M. and Shi J., A clinicopathologic and molecular update of pancreatic neuroendocrine neoplasms with a focus on the new world health organization classification, 143 (11): 1317-1326. © 2019 (59).

Imaging

 

Regardless of whether a PNEN is functional or non-functional, imaging is critical to assess the extent of the disease by localizing the primary tumor and identifying the size of metastatic disease. Localization is required preoperatively to increase the accuracy of intraoperative techniques and to reduce the need for repeated surgery. Besides, imaging is involved in patient’s management as it allows to monitor tumor growth and evaluate response to treatment (2, 25, 53, 55, 61). A multimodal approach is applied to diagnose and stage PNENs which comprises both anatomical and functional imaging modalities (2, 61-66).

 

ANATOMIC IMAGING   

 

Anatomical imaging modalities such as Computed Tomography (CT) and Magnetic Resonance Imaging (MRI) are capable of depicting normal and diseased tissue at high spatial resolution (67). Contrast enhanced CT is the most commonly used and preferred modality as it is widely available, renders clear anatomical images of the pancreas, lymph nodes, and liver metastasis and allows to assess vascular invasion and resectability (25, 52, 62, 64, 68, 69). The more recent, multiphase multidetector CT scan exhibits even more advantages including reduced artifacts due to rapid scan time, improved arterial phase images due to accurate contrast medium tracking and improved resolution by generating thinner slices that can be studied in different anatomical planes (61, 65). In addition, the frequent hypervascular nature of PNENs results in typical high contrast uptake in the arterial phase on CT which can aid in differentiating from pancreatic adenocarcinoma. The average sensitivity of contrast-enhanced CT varies from 63% to 83% and detection rates range from 69% to 94.3% (52, 67, 70). It appears that imaging of PNENs is often influenced by their biological heterogeneity. For example, gastrinoma become more apparent on postcontrast images and large NF-PNENs often have a necrotic or cystic appearance which tend to complicate diagnosis with imaging alone. In the latter case, MRI can be useful as cystic neoplasms can be better visualized due to the higher resolution, rendering MRI complementary to CT. MRI displays a similar sensitivity to CT (79%), but has some advantages over CT as it displays a good sensitivity even without administration of contrast agents, employs non-ionizing radiation, and is hence safer for patient follow-up (52, 53, 67). Limitations on the other hand include a higher frequency of motion-related artifacts as well as a longer acquisition time compared to CT (71).

 

Still, both conventional imaging modalities depend to a large extent on the tumor size (2, 25, 61, 72, 73). More than 70% of PNENs larger than 3 cm are detected, but only 50% of PNENs smaller than 1 cm are identified. As a result, small primary PNENs, especially insulinoma and duodenal gastrinoma, are frequently missed as well as small liver metastases (2, 25, 61, 62, 72-74). For small PNENs, which cannot be detected using CT and MRI, EUS is considered the predominant imaging technique (68). Because of the high spatial resolution of this modality, it is possible to localize even very small lesions (2-3 mm) (3, 74). Additionally, it is feasible to obtain high yield tissue samples by means of an FNA/B that can be used for Ki-67 measurements. Such EUS-FNA/B have a diagnostic accuracy of 80% for pancreatic adenocarcinoma and 46% for PNENs. In patients with proven insulinoma, EUS displays a sensitivity of 94% as a first-line modality. This renders EUS extremely valuable for localizing primary insulinoma (2, 68, 74, 75). However, EUS is not generally available, can be technically challenging, and results are operator dependent. In the hands of an expert, sensitivities of 79 to 100% can be achieved (61).  

 

FUNCTIONAL IMAGING  

 

Prior to the development of the current functional imaging modalities, selective angiography and sampling for hormone gradients were employed. However, due to the highly invasive nature of these techniques, minimally invasive modalities were developed which had a great impact on patient management (2, 53, 64).

 

Although PNENs exhibit highly heterogeneous biological behavior, 80-100% of PNENs, with the exception of insulinomas (50-70%), overexpress the G-linked protein somatostatin receptors (SSTRs), mainly subtypes SSTR-2 and -5. These receptors interact with somatostatin, a peptide hormone that affects neurotransmission and cell proliferation, but also the secretion of various compounds in the digestive system (2, 52, 62-64, 67, 76). Interestingly, these SSTRs also bind synthetic, radiolabeled somatostatin analogs (SSAs) with high affinity, which constitutes the basis of the primary functional imaging tool for PNENs, namely Somatostatin Receptor Imaging (SRI). SRI will not only allow to stage PNENs, but will also predictively identify patients eligible for SSA therapy (2, 52, 53, 64, 67, 68). One of the first SSAs used to target the SSTRs was octreotide labeled with 111Indium via chelator, diethylene-triamine-pentaaceticacid (DTPA). This 111In-DTPA-octreotide emits gamma rays that are detected 24 hours after intravenous injection using Single Photon Emission CT (SPECT) or SPECT/CT, the so-called Octreoscan® (53, 62, 64, 70, 77). The Octreoscan® is often combined with CT to improve the anatomic localization making it highly sensitive (77%), detecting 50-70% of primary PNENs, but less of the insulinomas and duodenal gastrinomas (2, 62-64, 68, 78-81). Major drawbacks include the availability and price of 111In-DTPA octreotide, the staggering acquisition time as well as the intrinsic shortcomings of SPECT, such as low spatial resolution (8-12 mm) (67, 82).

 

Positron Emission Tomography (PET) could provide better spatial resolution and greater precision (71). One of the most widely employed PET radiotracers currently used to image tumors is 18Fluor-labeled deoxy-glucose (18FDG). For high-grade NETs, especially NECs, 18FDG-PET/CT is a better choice as nuclear medicine modality as SSTR expression decreases when proliferation rates increase. 18FDG-PET can even be positive in G2 and G3 NETs. To this date, no cut-off value has been determined. However, it seems like neoplasms with Ki-67 values > 15% are more likely to exhibit a positive 18FDG-PET/CT, which is also a predictor of a more aggressive course (53, 81, 83, 84). However, 18FDG-PET/CT appears to be less useful in the majority of PNENs as these often show limited glucose uptake due to a rather slow growth rate (52, 64).

 

The development of new PET/CT radiotracers has been a major breakthrough in PNEN imaging. 11Carbon-5-hydroxytryptophan-labeled or 68Gallium-labeled SSAs including DOTA-tyrosine-3-octreotide (DOTA-TOC), DOTA-octreotate (DOTA-TATE), and DOTA-1-NaI-octreotide (DOTA-NOC) showed better sensitivity and diagnostic accuracy than the conventional imaging studies (Figure 4) and the Octreoscan® (Figure 5) (2, 52, 64, 71, 85-87). A meta-analysis revealed that 68Ga-DOTA-SSA PET for the diagnosis of NETs has a pooled sensitivity and specificity of 93% and 91%, respectively (88). Admittedly, the majority of the studies involved heterogenous populations, but most included a sizable minority of 20-30% PNENs. Hence, the overall data, although far from perfect, support use of 68Ga-DOTA-SSA PET over the Octreoscan® (89). Moreover, it is highly sensitive for the detection of bony metastases and it might obviate the need for additional radiologic studies. In addition to a higher sensitivity, other advantages of 68Ga-DOTA-SSA PET include patient convenience (imaging sessions take 70-90 minutes instead of 24 hours), lower radiation exposure, utility in finding unknown primary PNENs and it can lead to changes in treatment plans in about 33-41% of the patients (67, 89-93). 68Ga-DOTA-SSA PET might also be better at quantifying SSTR expression which facilitates targeted therapy such as PRRT (89). Consequently, 68Ga-DOTA-SSA PET quickly became the imaging modality of choice (67, 68, 71). However, similar to other imaging studies, false positives may occur due to pancreatic uncinate process activity, inflammation, osteoblastic activity, and splenosis (94). No doubt other PET agents will follow since PNENs express a variety of receptors for which there are potential ligands. For example, insulinomas express SSTRs in 50% of the cases, so tracers targeting the glucagon-like peptide-1 (GLP-1) receptor might be more useful in those patients (80, 95, 96).

Figure 4. Overview of (A) 68Ga-PET/CT, multiphase (B) atrial and (C) portal vein CT scan images from patient with partially cystic PNEN. The arrow indicates a liver metastasis which is only visible on the 68Ga-PET/CT scan. This figure has been adapted from Current treatment options in oncology, Morse B., Al-Toubah T. and Montilla-Soler J., Anatomic and functional imaging of neuroendocrine tumors, 21 (9): 75 © 2020 (67).

Figure 5. Comparison of (A) planar Octreoscan®, (B) Octreoscan®/SPECT/CT fusion, (C) planar 68Ga-DOTATOC-PET and (D) 68Ga-DOTATOC-PET/CT in the same patient. Images C and D clearly display a more precise delineation of the lesions. This figure has been adapted from International journal of endocrine oncology, Maxwell J. E. and Howe J. R., Imaging in neuroendocrine tumors: and update for the clinician, 2 (2): 159-68 © 2015 (82).

All benefits taken into account, the FDA approved 68Ga-DOTATOC PET in 2016 in the US, after being available in Europe for a number of years. Furthermore, with the development of an FDA approved 68Ga generator, an on-site cyclotron is no longer required, making this technology more widely available. A multi-society workgroup has recommended that 68Ga-DOTA-SSA PET replace use of Octreoscan®, unless it is not accessible, in combination with at least one anatomic imaging technique (66, 70).

 

Assessment Through Circulating Biomarkers

 

As stated earlier, the current gold standard for diagnosing and molecularly profiling PNENs remains the analysis of surgical or biopsy tissue samples. However, these samples have a highly invasive character, rendering repetitive sampling unfeasible. Further limitations are the individual patients’ risk and procedural costs. Besides, they represent merely a snapshot of tumor heterogeneity, which strongly influences accuracy. Hence, liquid biopsies aroused strong interest since they form a cost-effective and minimally invasive way to analyze the tumor’s behavior. The most frequently used source is blood as it allows to examine the so-called tumor circulome that consists of a set of circulating components that originate from the tumor (97-101). These blood-based biomarkers play a pivotal role in diagnosing and staging PNENs, monitoring response to therapy, and detecting tumor progression. In case of F-PNENs, specific circulating biomarkers such as insulin, gastrin, and glucagon are employed in hormonal assays to correctly diagnose F-PNENs. Moreover, both F- and NF-PNENs frequently secrete non-specific markers including CgA, neuron-specific enolase (NSE), pancreastatin, etc., which can be detected in patients’ blood as well (2, 9, 13, 52, 53, 55, 102). Besides circulating proteins, PNENs also shed circulating tumor cells (CTCs), circulating tumor RNA (ctRNA) and DNA (ctDNA) which could serve as potential biomarkers (98-100, 103, 104).

 

SPECIFIC BIOMARKERS AND HORMONAL ASSAYS  

 

Depending on the type of F-PNENs (outlined in Table 1), specific biochemical tests are performed. When insulinoma is suspected, serum levels of insulin and C-peptide are measured at a confirmed hypoglycemia during prolonged period of fasting (approximately 72 hours) as patients present with increased levels (> 6 µU/L and 0.6 ng/mL, respectively) even when glucose levels are low (7, 31, 52, 53, 104). In case of gastrinomas, the serum gastrin levels will be 10 times higher than the upper limit and gastric pH will be lower or equal to 2 (3, 7, 52, 53, 105). In patients with suspected VIPoma and glucagonoma, diagnosis is confirmed by determining the fasting levels of VIP and glucagon (7, 53).  

 

Chromogranin A (CgA)

 

CgA, a glycoprotein stored in and secreted by the secretory granules of the neuroendocrine cells, plays an important role not only in immunohistochemistry, but also as a circulating marker (7, 17, 52, 53, 102). CgA is useful as a marker for both functional and non-functional PNENs, as elevated levels are noted in 50-100% of the patients with PNENs (2, 106-108), depending upon the histological subtype (104, 109, 110). For example in gastrinoma, CgA levels are consistently high due to gastrin-induced enterochromaffin-like cell hyperplasia (111), while insulinoma show significantly lower levels of circulating CgA (112). Besides, blood levels depend upon malignant nature of the tumor, tumor burden, and progression, hence small tumors may be associated with normal CgA levels (113, 114).

 

The ENETS still recommends the use of circulating serum CgA as marker during diagnosis and follow-up in NF-PNENs (7, 115). However, the actual diagnostic value of this marker is still questionable (115). Sensitivity, specificity and overall accuracy of this clinical biomarker equal 66%, 95% and 71%, respectively (7), but these values tend to vary according to the specific assays and diagnostic threshold (52). Common conditions that can falsely elevate CgA levels, thus impair specificity, include decreased renal function, treatment with proton pump inhibitors (116), and even essential hypertension (117). In addition, 30-50% of NENs do not show elevated CgA levels, limiting sensitivity (47, 115, 118). This group mostly involves small, localized, non-functional NETs where CgA levels are normal in approximately 70% of the cases (119). As a result, these patients are in a higher need for accurate biochemical markers as diagnosis is harder both clinically and by use of imaging techniques. Moreover, SSA treatment cause a decrease in CgA secretion, which is why results should always be interpret with caution (104). In terms of follow-up, prospective studies demonstrated that elevated CgA levels do not correlate with imaging and tumor progression, hence questioning the potential of CgA as follow-up biomarker (115). Compared to CgA, CgB is not impacted by for example proton pump inhibitor treatment (112, 116). However, only in 25% of the cases with elevated CgA levels, CgB was elevated as well, thus routine estimation of CgB in all patients seemed not informatic in clinical practice (120).

 

Neuron-Specific Enolase (NSE)

 

NSE is a glycolytic enzyme expressed in the neuroendocrine cells of which levels can be elevated in PNEN patients, particularly those with a poorly-differentiated tumor (58, 110). However, its clinical use is limited as a blood-based biomarkers for NETs because sensitivity and specificity are only 39-43% and 65-73% to distinguish NETs from non-NETs. Consequently, NSE is therefore inferior to CgA in clinical practice (17, 110, 121). When combined with CgA measurement, sensitivity improves and reliability of NET diagnoses increases. Moreover, elevated CgA/NSE levels appear to provide prognostic information on progression-free survival (PFS) and OS (7, 102, 122).  

 

Pancreastatin

 

Pancreastatin, a post-translational processing product of CgA, is suggested to be a useful prognostic marker of NETs as pre-treatment levels > 500 pmol/L are an independent indicator of poor prognosis. Moreover, this marker is reported to correlate with the number of liver metastases and an increase in pancreastatin levels after treatment with SSAs is associated with poorer survival (52, 123). For diagnosis of NETs, pancreastatin is less sensitive than CgA, but also less susceptible to non-specific elevation (52, 121).

 

Pancreatic Polypeptide (PP)

 

PP is a hormone predominantly produced in pancreatic polypeptide cells, located in the head of the pancreas (7, 17, 109, 124). When used alone, a sensitivity of 63% is achieved in PNENs, but when combined with CgA sensitivity increases to 94%, better than either marker alone (109). However, less than 50% of PNEN patients display elevated serum levels and increases do not correlate with tumor burden and/or aggressiveness (102, 124). Moreover, there are several clinical conditions that can induce falsely elevated levels such as physical exercise, hypoglycemia, and food intake, whereas diarrhea, laxative abuse, high age, inflammatory processes and chronic renal disease could lead to a decrease (7, 102).   

 

Other Protein-Based Markers

 

Besides the above-mentioned markers, ProGRP and Neurokinin A can be used to further improve diagnostic and especially prognostic information. ProGRP in fact stimulates cell proliferation which is why increased levels are often associated with a more aggressive tumor and therefore worse prognosis (7, 12). In addition, several markers were reported to be useful for the detection of bone metastases that can be either osteolytic or osteoblastic. Bone alkaline phosphatase (BAP) indicates osteoblast function, while urinary N-telopeptide reflects osteoclast activity or bone resorption. An increased osteoclast activity predicts a poor outcome (12, 125, 126).    

 

Circulating Tumor Cells (CTCs)

 

CTCs have been investigated in a wide range of tumor types, and have gained increased interest in PNENs due to the limitations of the current circulating markers (98-100, 104, 121, 127, 128). The recently developed platform, CellSearch®, allows to detect and isolate CTCs based on expression of the epithelial cell adhesion molecule (EpCAM) on the cell membrane. EpCAM is a transmembrane epithelial glycoprotein that is overexpressed in adenocarcinoma, but recent studies (127-129) revealed EpCAM positivity in ileal, pancreatic, unknown primary, and gastric NETs as well. However, only 21-24% of the metastatic PNEN patients had detectable CTCs in the blood stream, which could potentially be explained by a slow shedding of CTCs or loss of EpCAM expression. Presence of CTCs was associated with increasing tumor burden and grade, while CgA failed to reveal this relationship. Changes in CTC levels were associated with treatment response and OS, revealing its potential as marker during treatment follow-up (127, 129). Furthermore, presence of CTCs could distinguish between patients suffering from PNENs with and without bone metastases with an area under the curve (AUC) of 79% (130). A phase II PAZONET study, during which Pazopanib treatment was evaluated, even demonstrated that patients without baseline CTCs showed improved response and longer median PFS (131). Contrarily, the CALM-NET phase IV study reported no notable effect of the presence of CTCs at baseline on PFS in patients treated with Lanreotide (132). Lastly, CTCs provide the opportunity to detect (epi)genetic alterations in PNENs through DNA and RNA extraction, but they can also be used to determine the SSTR status via immunohistochemistry which could facilitate therapeutic management (133, 134).

 

Circulating Tumor DNA (ctDNA)

 

ctDNA is the fraction of cell-free DNA (cfDNA) that originates from the tumor and constitutes one of the most promising new markers. It provides a representation of the whole tumor and contains tumor-specific genetic and epigenetic alterations, which allow to distinguish healthy from tumoral DNA (98-100, 104, 135). However, ctDNA research regarding NENs is still in its infancy. Boons et al., published the first paper confirming the presence of ctDNA in the plasma of metastatic PNEN patients by looking for tumor-specific single nucleotide variants (SNVs) via custom digital droplet PCR (ddPCR). In patients with localized PNENs, ctDNA could not be detected (136). In the same study, they revealed a significant correlation between CNA profiles of PNEN tissue and ctDNA and demonstrated the feasibility to detect ctDNA using these profiles (136). These findings were exploited in a more recent study, where they performed a cfDNA CNA analysis in a cohort of 43 NEN patients. Using this analysis, ctDNA could be detected in 13 of the 21 PNEN patients. ctDNA positivity appeared to be significantly associated with higher WHO grade, location of the primary tumor and higher levels of CgA and NSE. Besides, a worse OS was observed in ctDNA-positive patients. In addition, they illustrated that CNA patterns in cfDNA could even assist in distinguishing PNENs from the more common PAADs. Moreover, the longitudinal tumor fraction (i.e., amount of ctDNA vs. total cfDNA) measurements were associated with PFS and could indicate tumor progression (47).  

 

MicroRNA (miRNAs)

 

miRNAs are short noncoding RNAs (< 30 nucleotides) designated to regulate many processes including cell proliferation, apoptosis, and development (134, 137), by inducing translational repression or degradation of certain mRNAs (138). In cancer, miRNA regulation is often altered as is the case in PNENs (138). Normal pancreatic islets and PNENs display a distinctly different miRNA profile as PNENs express miRNA-103 and-107, but lack miRNA-155. A set of 10 miRNAs was even able to perfectly distinguish 40 PNENs from 4 PAADs (137). miRNA-204 was overexpressed in insulinomas only, miRNA-196a had a prognostic function and overexpression of miRNA-21 was associated with higher Ki-67 rates and presence of liver metastasis (137). A more recent study, demonstrated that the combination of a set of miRNAs together with CgA measurements could improve diagnostic accuracy (139). However, data on circulating miRNA is still scarce as miRNA measurements in NETs are not properly standardized, requiring further research (140).

 

NETest

 

The NETest, a blood-based multi-analyte transcript assay, was developed in 2013 by Modlin and colleagues (141). The expression of 51 marker genes, encompassing genes associated with NENs, is examined using a quantitative PCR (qPCR) and analyzed using multivariate algorithms (142, 143). These algorithms enable the calculation of a disease-activity score, ranging from 0 to 100, with scores higher than 20 representing tumoral samples (140, 142, 143). The NETest captures accurate diagnosis and tumor biology of NETs with the most recent study demonstrating an accuracy of more than 91%. More specifically, the NETest has proven useful for diagnosing PNEN patients, as PNENs could be distinguished from other pancreatic malignancies with an accuracy of 94% (142, 143). The test also shows to be a real-time monitor of clinical status through the disease-activity score of NEN patients. Low biological activity corresponds to a score of less than 40, while intermediate and high biological activity, indicating tumor progression, have scores of 41-79 and 80-100, respectively (103, 140, 143). Stable and progressive disease could be differentiated with an accuracy of 84.5-85.6%, consistent with image-based categorizations (103). Moreover, changes in NETest disease-activity scores over time correlated with response to treatments including SSAs, PRRT, and surgery (144-147). For example, in a prospective analysis, performed by Modlin and colleagues, 35 pancreatic and small intestine NEN patients were included that all displayed elevated NETest levels prior to surgery, while only 14 of them had increased CgA levels. After tumor removal, the disease-activity scores reduced from 80 ± 5 to 29 ± 5 (p < 0.0001), whereas changes in CgA levels did not correlate with resection. Four of the 11 patients with complete tumor resection still presented increased NETest scores one month after surgery and showed positive evidence of recurrence 6 months post-surgery (144).

 

Since 2013, the NETest has proven to perform better than the single analyte tests (e.g., CgA) and these results appeared to be highly robust and reproducible (103, 140, 142). However, a large independent validation study conducted in the Netherlands has revealed that the test is more sensitive, but less specific than CgA suggesting its suitability as a marker for disease follow-up, but not as a screening tool (147). This test is not affected by food intake or specific medication, is easy to use and available which all increases clinical utility (140). The NETest possess advantages from an economic point of view too. Identifying patients with molecularly stable disease (SD) could potentially lead to fewer use of imaging modalities. Moreover, by enabling faster identification of the clinical status than with imaging, ineffective therapies can be ceased more quickly with another obvious cost-benefit effect (140, 142). Despite all advantages, NETest is currently not implemented in a clinical setting. Results of additional independent validation studies and other practical aspects such as costs and transparency will ultimately determine its integration in clinical practice.

 

MANAGEMENT OF PNENs

 

With a better understanding of the heterogeneity in PNENs, the number of treatment options has increased substantially over the years. Unfortunately, there is a lack of head-to-head comparison data. Therefore, treatment must be individualized considering the age and overall health of the patient, the specific toxicities of potential treatment(s), costs, and potential impact on quality of life (QOL). Consequently, decisions with regard to patient management must be made by an experienced, multidisciplinary team together with the primary care physician (52). Generally, the management of PNEN patients consists of a series of well-defined steps. These comprise of: 1) establishing a diagnosis, 2) determining localization and extent of tumor, 3) controlling hormone excess state in case of F-PNENs, 4) resecting tumor, if possible, 5) checking for presence of hereditary disease (MEN1), 6) treating advanced and metastatic PNENs, and 7) long-term monitoring for tumor progression (63, 148).

Figure 6. Possible treatment scheme for PNENs based on functionality and extent of the tumor. This figure has been adapted from Cancers, Akirov A., Larouche V., Alshehri S. et al., Treatment options for pancreatic neuroendocrine tumors, 11 (6) © 2019 (149).

It is crucial to consider grade/differentiation, stage/extent, and functional status of the tumor as different treatment schemes evolved based on these factors (Figure 6). For example, surgery is usually advocated for PNENs that are functional, larger than 2 cm, or intermediate-to-high grade (3, 8, 52). For patients with metastatic disease, the treatment options are extensive and encompass surgical debulking, systemic therapies including chemotherapy, or targeted therapy such as liver-directed therapy and peptide receptor radionucleotide therapy (PRRT) (149). It is not unusual for the management plan to change based on treatment response and disease progression. Failure to respond to treatment or unexpected changes in the tempo of disease due to tumor dedifferentiation and tumor heterogeneity are well-described in PNENs. Accordingly, most patients will receive multiple treatments during the course of their disease, but there is no data on the optimal treatment sequence (52, 105). The various treatment modalities are discussed below.

 

Surgical Management

 

Surgery continues to play a major role in the management of patients with PNENs as it remains the only potentially curative treatment for PNEN patients and it can alleviate clinical symptoms caused by excessive hormone production and tumor bulk (2, 3, 7, 149-151). Furthermore, several studies revealed that patients who underwent surgical resection had a reduced risk of metastases as well as showed an improved disease-free survival (DFS) (152, 153). Different approaches exist such as resection of the primary tumor and surrounding lymph node metastases through pancreaticoduodenectomy (Whipple procedure) and pancreatectomy (central or distal) as well as the more conservative methods including sparing enucleation and wait-and-see observations (7, 52).

 

Choosing the appropriate approach depends on the extent and location of the tumor, the functional status as well as the presence or absence of metastases (52, 150, 154-156). Generally, surgery is recommended in patients with localized NF-PNENs. Besides the primary tumor, peritumoral metastases should be eradicated as well since nodal metastases occur in at least 30% of NF-PNEN patients which affect tumor grade, but more importantly DFS (157, 158). Exceptions occur in patients with sporadic, low-grade (G1/G2) NF-PNENs smaller than 2 cm (3, 8, 52, 149, 150, 154-156). For those patients, optimal management is controversial as some recommend surgical interventions such as enucleation, whereas others including the ENETS advocate a wait-and-see attitude due to the indolent nature of these tumors (3, 8, 52, 148-150, 159, 160). A similar conservative approach is encouraged in MEN1 patients with NF-PNENs of 2 cm or smaller as these tend to have a low disease-specific mortality (161). Thakker and colleagues, on the other hand, suggest resection of NF-PNENs larger than 1 cm that demonstrate significant growth over 6 to 12 months (162).

Surgical excision of the tumor is also recommended for patients with F-PNENs as these display high cure rates (2, 13, 149, 163-165). The National Comprehensive Cancer Network (NCCN) guidelines describe that insulinomas and gastrinomas are preferably removed by enucleation with peritumoral node dissection if the tumor is located in the head of the pancreas. Deeper, more invasive tumors are more appropriately eradicated by pancreaticoduodenectomy. The former strategy should also be applied for small peripheral glucagonomas and VIPomas. PNENs in the distal part of the pancreas, in turn, are ideally removed through distal pancreatectomy (149). Surgery for MEN1 patients with NF-PNENs and gastrinoma remains controversial as they often present with multiple primary tumors which renders curative surgery almost impossible. Aggressive resection of all PNENs smaller than 2 cm in MEN1 patients seems contra-indicated as several studies revealed that these patients rarely develop advanced disease and have a good prognosis (2, 18, 75, 165-167).

 

The traditional surgical approach is open laparotomy as this allows thorough abdominal exploration including bimanual palpation and intraoperative ultrasound of the pancreas and liver (2, 168). However, several studies reported that certain lesions in particular those amenable to enucleation or to distal pancreatectomy may be approached with laparoscopic or robotic techniques (169). Venkat and colleagues even demonstrated that patients who underwent laparoscopic resection had less blood loss and a lower overall complication rate, and were consequently permitted to leave the hospital sooner than patients who had had open pancreatic resection (170). Gastrinomas form an exception since palpation plays an important role in the detection of these often small malignancies. Moreover, 60-90% of these patients will have lymph node metastases in addition to the primary tumor (169, 171). Adopting a purely laparoscopic approach to these tumors will depend upon improvements in haptic feedback technology. For tumors requiring a Whipple procedure both laparotomic and laparoscopic approaches are used in centers worldwide as the latter is still associated with technical difficulties. However, when performed by trained hepatobiliary or laparoscopic surgeons’ effectiveness and safety are similar and, in some cases, even superior to open surgery (168, 171).     

 

In patients with distant metastases, surgical intervention remains important, although it may no longer result in cure (52). The most common site of distant metastasis is the liver since 46-93% of NET patients develop liver metastases which can lead to liver failure, a common cause of death (52, 172-174). There are multiple options available for patients with hepatic metastases, including surgical resection which, in selected cases, appears to improve survival in uncontrolled series (157). The optimal approach depends on several factors including the extent of primary tumor and liver metastases, planned treatment as well as the age and overall health of the patient. Accordingly, the NCCN recommends complete resection (R0 resection) of primary tumor and liver metastases, if possible and otherwise consider tumor debulking (149). Aggressive resection of the primary tumor in the setting of liver metastases is associated with a survival benefit as both obstruction and further metastatic spread may be prevented. The 5-year survival rate after this surgery ranged from 65% to 73% which is significantly better than that of patients with nonresectable metastases (20%) although this difference might be at least partially explained by selection bias, where only very fit patients receive surgery (174-176). In case of the latter, numerous non-surgical options are available (see liver-directed therapy) and primary tumor, when found to be asymptomatic and stable, is not removed (52, 177). However, R0 resection can only be achieved in 10-20% of the cases as the majority of patients presents with multifocal and bilateral metastases and studies suggest that only one third of all liver metastasis are visible on imaging (52, 174, 175, 178, 179). Consequently, cytoreductive hepatic surgery is more frequently opted for, but this approach remains controversial as it is incomplete and the target population is not clearly described. It is therefore generally considered that patients with metastatic G1/G2 PNEN in which preferably less than 25% of the liver is affected are eligible for tumor debulking (2, 52). Several studies already showed that this procedure can alleviate clinical symptoms in F-PNENs, but also provide better long-term survival (2, 52, 149, 180-183). Moreover, debulking may also be associated with an improved response to concomitant therapy such as embolization (184). Radiofrequency ablation (RFA) is increasingly used in PNEN patients to address hepatic metastases and is often performed during surgery or laparoscopically (2). Patients with extensive liver involvement, who are consequently ineligible for R0 resection and tumor debulking, may be aided with a liver transplant to improve life expectancy (52, 176, 185). A non-randomized study in 88 patients who met strict criteria of transplant eligibility reported a difference in OS in the transplant (88.8%) and no transplant group (22.4%) after 10 years (185). Important concerns are the availability of the grafts as well as the lifelong immunosuppression required after transplantation. Also, the exact criteria for eligibility are very similar to those for tumor debulking which makes patient selection difficult (52).

 

Medical Therapy

 

Use of medical therapy is limited to those with locally advanced or metastatic disease. Some of the current and promising options for targeted systemic therapy are shown in Figure 7.

Figure 7. Overview of the current (blue) and promising (red) options for targeted medical therapy in (P)NETs. This figure has been retrieved from Drugs, Herrera-Martinez A. D., Hofland J., Hofland L. J., Targeted Systemic Treatment of Neuroendocrine Tumors: Current Options and Future Perspectives 79:21–42 © 2019 (186).

Figure 8. Visualization binding affinity of each of the three FDA-approved SSAs to the different SSTR subtypes. This figure was retrieved and adapted from Drugs, Herrera-Martinez A. D., Hofland J., Hofland L. J., et al., Targeted systemic treatment of neuroendocrine tumors: current options and future perspectives, 79:21-42. © 2019 (186).

SOMATOSTATIN ANALOGS  

 

As previously described in the functional imaging section, SSTRs are often highly overexpressed in PNENs. Several SSAs including Octreotide (Sandostatin®) and Lanreotide (Somatuline®), which have affinity for different SSTR subtypes (Figure 8), were therefore marketed. These inhibit hormone secretion and thus reduce clinical symptoms in patients with F-PNENs (149, 186). Additionally, several studies revealed that SSAs are also capable to control tumor growth with a positive impact on PFS. The antiproliferative effect of SSAs in PNETs was confirmed in the Controlled Study of Lanreotide Antiproliferative Response in Neuroendocrine Tumors (CLARINET) trial. A total of 204 patients with well-differentiated, progressive NETs were included who were then randomly assigned to either Lanreotide or placebo treatment for 96 weeks. After 24 weeks, PFS rates were 65.1% and 33.0% in the Lanreotide and placebo groups respectively (Figure 9). The study also demonstrated that there was no significant difference in QOL and OS in both groups (149, 186-189).

Figure 9. PFS among patients that received Lanreotide (red) or placebo (blue). This figure was retrieved and adapted from The New England journal of medicine, Caplin M. E., Pavel M., Cwikla J. B., et al., Lanreotide in metastatic enteropancreatic neuroendocrine tumors, 371 (3): 224-33 © 2014 (189).

As an extension to the core CLARINET study, the CLARINET open-label extension (OLE) reported long-term safety and additional efficacy data. For this purpose, 88 patients with SD were selected from the core study. Forty-one patients continued their Lanreotide treatment, while 47 patients shifted from placebo to Lanreotide. Safety and tolerability were favorable during a mean treatment period of 40 months. In addition, adverse effects, that were either attributable or not to Lanreotide, were found to improve as duration of treatment, hence exposure, increased. Median PFS in patients who had already received Lanreotide in the core study was estimated at 32.8 months (Figure 10). Of the 32 placebo-treated patients who exhibited progressive disease (PD) in the core study, 17 patients persisted in PD, while the remaining 15 patients had a median time to progression (TTP) of 14 months (187, 190). Based on the findings from the CLARINET trial, the use of SSAs as first-line treatment for symptom relief and tumor control is recommended in the NCCN and ENETS guidelines for advanced, well-differentiated, unresectable PNENs, particularly those with a high burden of liver metastases (149, 188, 191).

Figure 10. PFS of patients that received Lanreotide in the core and OLE CLARINET study. The OLE data is only visualized for patients that were originally assigned to and continued the Lanreotide treatment. This figure was retrieved and adapted from Endocrine-related cancer, Caplin M. E., Pavel M., Cwikla J. B., et al., Anti-tumor effects of lanreotide for pancreatic and intestinal neuroendocrine tumours: the CLARINET open-label extension study, 23 (3): 191-9 © 2016 (190).

MOLECULAR-TARGETES AGENTS  

 

Newly developed molecular-targeted treatments including Sunitinib and Everolimus (Figure 7) have shown to improve PFS in advanced, metastatic PNENs and represent the most common second line treatments that are currently available (52, 149). An overview of the most recent findings can be found in Table 3.    

 

The tyrosine kinase inhibitor (TKI), Sunitinib, has been approved for the treatment of patients with well-differentiated, unresectable, locally advanced or metastatic PNENs as it displays an antiangiogenic working mechanism. It actually inhibits vascular endothelial growth factor receptors (VEGFR) 1 and 3, stem cell factor (SCF) receptor as well as platelet-derived growth factor receptors (149, 186, 192). A two-cohort phase II study, examining 107 patients with advanced NETs (of which 66 PNENs), reported an overall objective response rate (ORR) of 16.7% and SD in 68% of PNEN patients. Median TTP was 7.7 months in PNENs and 10.2 months in carcinoid patients (193). A phase III multinational, randomized, double-blind, placebo-controlled trial (SUN 1111) confirmed the activity of Sunitinib in patients with advanced, well-differentiated PNENs (Figure 11A). A total of 171 patients were enrolled in this study. Median PFS was 11.4 months in the Sunitinib group compared to 5.5 months in the placebo group, with the latter group having a higher mortality rate (25% vs 10%) (194). A retrospective analysis of the previous phase III trial reported an increased PFS in both the Sunitinib and placebo group (12.6 vs. 5.8 months). Median OS after 5 years were 38.6 and 29.1 months of the Sunitinib and placebo groups, respectively. Important to note here is that 69% of the placebo-treated patients shifted to Sunitinib treatment (195). Sunitinib presented with an acceptable safety profile as the most frequent adverse effects in the sunitinib group included diarrhea, nausea, vomiting, asthenia, and fatigue which can be managed through dose interruption or modification (192, 194, 196).  

 

Everolimus (Afinitor®) is an oral, protein kinase inhibitor of the mammalian target of rapamycin (mTOR) pathway that displays proven antitumor activity in advanced PNENs, either alone or combined with Octreotide therapy. A multinational phase II study, the RADIANT 1 trial, has reported the efficacy of Everolimus alone and in combination with Octreotide in patients with metastatic PNENs that have progressed on chemotherapy (197). Monotherapy with Everolimus provided SD in 67.8% of patients and a partial response (PR) in 9.6%, while combination therapy resulted in 80% SD and 4.4% PR. Everolimus treatment also led to a decrease in CgA and NSE levels in 50.7% and 68.2% of the patients (Table 4). An early tumor marker response (i.e., > 50% decrease by 4 weeks) was associated with a significantly longer PFS (197). The RADIANT 3 trial, later on, investigated Everolimus as first line therapy in patients with advanced PNENs (Figure11B). Four hundred and ten patients with radiologic progression of disease were randomized to either Everolimus (10 mg once daily) or placebo. The median PFS was 11 months with Everolimus compared to 4.6 months with placebo representing a 65% reduction in estimated risk of progression or death. The proportion of patients alive and progression free at 18 months was 34% with Everolimus compared to 9% with placebo. Toxicities were mostly grade I or II (198). Similar PFS rates were reported regardless of whether patients were chemo-naïve or had received prior chemotherapy (199, 200). Addition of Pasireotide to Everolimus did not improve PFS compared to Everolimus alone (201).

 

Based on the recent, above-mentioned data, the European Society for Medical Oncology (EMSO) guidelines 2020 recommend the use of molecular-targeted agents such as Sunitinib and Everolimus in advanced, progressive PNENs (G1/G2) (191). Likewise, the North American Neuroendocrine Tumor Society (NANETS) guidelines 2020 recommend both treatments for well-differentiated, metastatic PNETs (G1/G2) (202). Both guidelines state there is no support to use Sunitinib nor Everolimus in treatment of PNET G3 or PNECs (191, 202). When comparing both molecular-targeted agents, response rates appear comparable (Figure 11). Since there has been no trial comparing the two agents directly, choice of agent may be based on the potential side-effects and patient’s overall health. For example, in patients with poorly-controlled hormonal symptoms, especially hyperinsulinism, congestive heart failure, hypertension, high risk of gastrointestinal bleeding or a history of myocardial infarction or stroke, Everolimus is thought to be the preferred choice (194, 202, 203). On the other hand, in patients with poorly controlled diabetes mellitus, pulmonary disease, or high risk of infection, sunitinib would be a more appropriate choice (192, 203). Moreover, up until now several biomarkers have been identified that correlated with the patient’s outcome. An overview of the currently known biomarkers can be found in Table 4 (204).

 

 Table 3. Results from Most Important Phase II and III Studies of Sunitinib and Everolimus in PNENs

Study

Patients

Active treatment

PD at entry

ORR

PFS/TTP (months)

Safety and other comments

Sunitinib

Phase II, open label (193)

N = 107 

 

- PNETs = 66

 

 

- Carcinoid = 41

 

50 mg daily

Schedule 4/2*

No

 

 

ORR = 16.7%

SD = 68%

 

ORR = 2.4%

SD = 83%

 

 

TTP = 7.7

 

 

TTP = 10.2

G3 fatigue: 24.3%

Phase III,

RCT,

SUN 1111 (194)

 

[Retrospect]

(195)

N = 171

 

- Sunitinib = 86

 

 

 

- Placebo = 85

 

 

37.5 mg daily

CDD**

Yes

 

 

ORR = 9.3%

SD = 63%

PD = 14%

 

ORR = 0%

SD = 60%

PD = 27%

 

 

PFS = 11.4

[PFS = 12.6]

 

 

PFS = 5.5

[PFS = 5.8]

Common AEs:

30%: diarrhea, nausea, asthenia, vomiting and fatigue 

 

10-12%: G3/4 neutropenia and hypertension

Everolimus

Phase II,

open label,

RADIANT 1 (197)

N = 160

 

- Stratum1 = 155

 

 

 

- Stratum2 = 45

 

 

10 mg daily

 

 

 

10 mg daily + 30 mg LAR Octreotide

Yes

 

 

PR = 9.6%

SD = 67.8%

PD = 13.9%

 

PR = 4.4%

SD = 80%

PD = 0%

 

 

PFS = 9.7

 

 

 

PFS = 16.7

 

Specific AEs:

 

5.2%: G3/4 asthenia

 

 

8.9%: G3/4 thrombocytopenia

 

Common AEs:

30%: stomatitis, rash, diarrhea, fatigue, nausea

Phase III,

RCT,

RADIANT 3 (198)

N = 410

 

- Everolimus = 207

 

 

- Placebo = 203

10 mg daily

Yes

 

 

PR = 5%

SD = 73%

 

PR = 2%

SD = 51%

 

 

PFS = 11

 

 

PFS = 4.6

 

Common AEs:

64%: stomatitis

49%: rash

34%: diarrhea

31%: fatigue

23%: infections

Abbreviations: ORR, objective response rate; PFS, progression-free survival; TTP, time to progression; SD, stable disease; PD, progressive disease; CDD, continuous daily dosing; AE, adverse event; LAR, long-acting release; PR, partial response; RCT, randomized clinical trial

* Concomitant use of SSA in 27% of PNET patients and 54% of patients with carcinoid tumors.

** Concomitant use of SSA in 26.7% of patients.  

 

Figure 11. This figure compares the PFS in patients with advanced metastatic PNENs, (A) treated with Sunitinib in the SUN 1111 trial (194) and (B) Everolimus in the RADIANT 3 trial (198). Figure A was retrieved and adapted from The New England journal of medicine, Raymond E., Dahan L., Raoul J. L., et al., Sunitinib malate for the treatment of pancreatic neuroendocrine tumors, 364 (6): 501-13 © 2011 (194). Figure B was retrieved and adapted from The New England journal of medicine, Yao J. C., Shah M. H., Ito T., et al., Everolimus for advanced pancreatic neuroendocrine tumors, 364 (6): 514-23 © 2012 (198).

 

Table 4. Current Soluble Biomarkers and Correlations with Outcomes with Targeted Therapies in PNENs

Study

Biomarker

Results

Sunitinib

(204, 205)

 

VEGF

Increased in 53% of patients after 4 weeks of treatment

Return to baseline after 2 weeks off treatment

When Sunitinib level > 50 ng/dL higher changes observed

No significant difference between PNET and carcinoids

sVEGFR

Decrease of ³ 30% in sVEGFR-2 and -3 levels

Return to baseline after 2 weeks off treatment

Reduction in sVEGFR-3 correlated with better OR and PFS

Lower baseline sVEGFR-2 with radiological SD for > 6 months

Elevated baseline sVEGFR-2 correlated with improved OS

IL-8

Increase (>2-fold) in 43% and (>3-fold) in 23% of patients after 4 weeks of treatment

Return to baseline after 2 weeks off treatment

Increase (1.8-fold) after 4 weeks on treatment

SDF-1a

Increase (20%) after 4 weeks on treatment

Elevated baseline correlated with significantly shorter TTP, PFS and OS

Everolimus

(197, 206)

 

CgA

Increase (> 2-fold) of CgA at baseline correlated with decreased PFS and OS

Reduction of > 30% in CgA levels after 4 weeks correlated with increased PFS and OS

NSE

Elevated NSE levels at baseline correlated with decreased PFS and OS

Reduction of > 30% in NSE levels after 4 weeks correlated with improved PFS

Abbreviations: VEGF, vascular endothelial growth factor; sVEGFR, soluble VEGFR; SDF-1a, stromal cell-derived factor 1 alpha.

Note: This table was adapted from Molecular Diagnosis and Therapy, Mateo, J., Heymach, J. V. and Zurita, A. J., Biomarkers of response to Sunitinib in gastroenteropancreatic neuroendocrine tumors: current data and clinical outlook, 151-161. © 2012 (204).

 

CYTOTOXIC CHEMOTHERAPY  

 

There is currently no unanimity on which cytotoxic chemotherapy would be optimal for the treatment of PNENs. Therefore, patient selection is key so factors such as primary tumor site and stage, differentiation, and proliferation index should be considered. Conventional cytotoxic chemotherapy is often used as first-line therapy for metastatic and progressive PNETs or PNECs (149, 207). ENETS guidelines describe the following indications: progression under SSA treatment, worsening clinical symptoms, and/or Ki-67 values > 10% (208). In a neoadjuvant setting, chemotherapy can play a potential role in tumor shrinkage prior to resection (7). Two major types of chemotherapeutic agents can be distinguished namely alkylating and platinum agents (7, 149, 207). In practice these are often combined with antimetabolites and anthracyclines (209). An overview of the most commonly used combinatory therapies and when to employ them is described in more detail below.

 

Alkylating agents such as Streptozocin, Dacarbazine, and Temozolomide are key in the treatment strategy of PNEN patients since they are often employed as second line treatment after progression under SSA (207). First of all, Streptozocin, a nitrosourea alkylating agent, is taken up by cells via a glucose protein 2 (GLUT2) after which cell damage is induced. Since the compound is associated with significant renal and hematological toxicity in high doses, it is often combined with 5-fluorouracil (5-FU) or Doxorubicin with dose reduction, hence reduced toxicity as a result (2, 149, 209). A comparative, phase III study conducted in 1992 reported that the combinatory therapy of Streptozocin + Doxorubicin provided more favorable results than Streptozocin + 5-FU in patients with advanced PNENs (210). However, the results described in this study have not been confirmed in any subsequent study (163, 209). Kouvaraki and colleagues retrospectively studied 84 PNEC patients treated with Streptozocin, 5-FU and Doxorubicin. Response rate was 39% with 2-year PFS and OS of 41% and 74%, respectively (211). Dacarbazine, a second alkylating agent, serves as a less toxic alternative. A phase II study tested Dacarbazine as a monotherapy in 50 PNEN patients and reported an ORR of 34% and median OS of 19.3 months (212). When combined with 5-FU, the overall response rate and PFS in advanced NENs were 38.2% and 13.9 months, respectively (213). A third alkylating agent that is primarily used as monotherapy for treatment of glioblastoma and melanoma is Temozolomide (163, 209). When combined with other compounds including Capecitabine (214), Bevacizumab (215), Bevacizumab and Octreotide (216), Thalidomide (217) and Everolimus (200) it shows significant activity in advanced PNENs (149, 209). A 2011 retrospective study reported that the combination treatment Capecitabine + Temozolomide (CAPTEM) in 30 chemonaive NEC patients resulted in an ORR of 70% with a PFS of 18 months (214). In 2018, a prospective, randomized phase II trial investigated Temozolomide therapy versus the CAPTEM combination therapy in PNEN patients. PFS was significantly better in the latter group (14.4 vs. 22.7 months) (218). However, a more recent retrospective analysis showed that CAPTEM was not able to improve PFS. Consequently, it was suggested by the authors that CAPTEM might be more useful for tumor shrinkage rather than improving PFS (219). 

 

In poorly-differentiated G3 NECs, platinum agent regimens are often used in patients with adequate performance status. The first-line chemotherapy for NEC patients encompasses Cisplatin or Carboplatin combined with Etoposide or Irinotecan, based on the reported results (52, 220-223). Moertel and colleagues investigated the effect of Cisplatin + Etoposide in 45 patients with metastatic NENs, of which 27 were well-differentiated. The ORR was 67% in the 18 poorly-differentiated NECs with complete response (CR) in 17% of the patients, while unfortunately, only 2 patients (17%) of the well-differentiated NEN patients showed a response. Moreover, they reported a median survival of 19 months which was significantly longer than those described in literature (6-7 months). However, toxicity was a major issue (220). These results were confirmed by Mitry and colleagues in 1999 (221). Lower toxicity levels were observed when patients were treated with Carboplatin, but efficacy was similar to that of Cisplatin, rendering Carboplatin a valuable alternative (222, 224). Moreover, Oxaliplatin-based therapy appeared to have a greater activity in advanced PNETs (207).

 

The role of second-line chemotherapy for NEC patients is currently unknown, but many combinatory options have been examined (223, 225). Capecitabine + Oxaliplatin (CAPOX) and 5-FU + Oxaliplatin (FOLFOX) have been evaluated in two retrospective trials in well-differentiated NENs. ORRs of 26% and 30% were reported, respectively (226, 227). In addition, FOLFIRI and FOLFIRINOX (5-FU-based chemotherapies) have recently proven some effect in NEC patients progressing on platinum-based regimens (225, 228).

 

Radiotherapy

 

PEPTIDE RECEPTOR RADIONUCLIDE THERAPY (PRRT)

 

Peptide receptor radionuclide therapy (PRRT) is a therapy whereby a radiolabeled SSA (117Lutetium or 90Yttrium) is used to treat SSTR-positive, locally advanced and/or metastatic GEP-NENs, including PNENs. Adverse effects include nausea, renal toxicity, transient bone marrow suppression and seldom myelodysplastic syndrome or acute myeloid leukemia in 1-2% of patients (7, 149, 229, 230).

 

In a study of 504 patients, treatment with the analog 177Lu-DOTATATE showed activity in GEP-NENS (230). Looking specifically at the PNEN subgroup there was a 6% CR and a 36% PR in NF-PNENs and no CR and 47% PRs in functioning PNENs (230). Striking improvements in QOL of responders was also noted (231). A more recent study of 68 patients with PNENs treated with PRRT showed PRs in 41 patients (60.3%), minor responses in 8 (11.8%), SD in 9 (13.2%) and PD in 10 (14.7%) (232). The authors concluded that the outstanding response rates and survival outcomes suggest that PRRT is highly effective in advanced G1/2 PNENs when compared to other treatment modalities. Independent predictors of survival were the tumor proliferation index, the patient’s performance status, tumor burden and baseline plasma NSE level. PRRT also provided improvements in PFS compared to Octreotide in midgut NENs (232). The NETTER-1 phase III trial confirmed the efficacy in PRRT in midgut NENs in 2017 (229, 233, 234). Therefore, the FDA approved use of 177Lu-DOTATATE based on the results obtained in the NETTER-1 trial in midgut NETs (229, 234). Thus, the number of centers where this treatment is available is expected to increase in the US, although it has been used in Europe since 1996. Joint society practice guidelines have been developed (235). There are a number of ongoing international clinical trials listed on Clinical Trials.gov. Third party payer reimbursement is an ongoing issue which hopefully will be resolved.

 

For PNENs, the effects of PRRT have only been investigated in several single-arm prospective and retrospective trials (229). These, however, identified several signals in favor of PRRT use in PNENs as disease control rates and PFS varied between 84-85% and 30-34 months, respectively (232, 236). Additionally, a meta-analysis compared the efficiency of PRRT to Everolimus in GEP-NENs that were not eligible to surgical resection. An ORR of 47% was reported in the PRRT treated subgroup versus only 12% of the Everolimus treated patients. Moreover, disease control rates (81% vs. 73%) as well as PFS (25.7 vs. 14.7 months) were also superior in the PRRT treated subgroup (237). A recent retrospective study evaluated the association of upfront PRRT vs. upfront chemotherapy or targeted therapy with PFS in enteropancreatic NET patients who progressed under SSA treatment. Patients with a Ki-67 value of £10% who received upfront PRRT, were reported to have a statically and clinically meaningful prolonged PFS (238). Based on these findings, it seems important to better define the role of PRRT in the treatment of PNENs within the future.

 

Liver-Directed Therapy

 

As mentioned earlier, the liver constitutes the most common site for distant metastases (52, 172-174). Since surgical resection and RFA are only feasible in a minority of patients, there are multiple liver-directed therapies available to treat the remaining patients. These methods include transarterial chemoembolization (TACE), transarterial embolization (TAE), or radioembolization, which will be discussed below. Given the lack of randomized data, it is difficult to determine with certainty which method is preferred. Moreover, NANETS guidelines recommend to consider systemic therapy rather than liver-directed therapy when >50-75% hepatic tumor burden is present (239).  

 

A study of chemoembolization combined with SSA treatment resulted in a relief of symptoms in 78% of the patients. Monitoring serum pancreastatin levels predicted a response to this therapy in which radiographic improvement or stability were seen in 45% of patients (77). In NEN patients that underwent TACE, plasma levels of pancreastatin above 5000 pg/ml pre-treatment were associated with increased peri-procedure mortality (240).

Radioembolization (also known as selective intrahepatic radiotherapy or SIRT) involves embolization of 90Yttrium embedded either in a resin microsphere (Sir-Sphere) or a glass microsphere (TheraSphere). Acute toxicities associated with 90Yttrium microsphere embolization appear to be lower than other embolization techniques, primarily due to the fact that the procedure does not induce ischemic hepatitis. Thus, the procedure can be performed on an outpatient basis. A rare, but potentially serious complication is radiation enteritis, which can occur if particles are accidentally infused into arteries supplying the gastrointestinal tract. Chronic radiation hepatitis is another potential toxicity. Response rates associated with radioembolization in metastatic NEN patients have been encouraging. In one retrospective multi-center study of 148 patients treated with Sir-Spheres, the objective radiographic response rate was 63% with a median survival of 70 months, with no radiation-induced liver failure (241). Another study of 42 patients treated with either Sir-Spheres or TheraSpheres reported a response rate of 51%. However, only 29 of the 42 enrolled patients were evaluable (242). Grozinsky-Glasberg and colleagues examined 57 patients which underwent either TACE, TAE or SIRT. They reported symptomatic control and a stabilization of tumor growth in 95% of the patients. Noteworthy, they observed improvements regardless of the extent of the liver metastasis (243).

 

Novel Targets for the Treatment of (P)NENs

 

While there has been a quantum leap in the ability to treat NENs successfully we have a long way to go to cure the disease. Fortunately, research into improved and novel therapeutic strategies is ongoing. So far, the results of immunotherapy as monotherapy in PNET patients remain disappointing. Examples include the inhibition of the programmed death-ligand 1 (PDL-1) and cytotoxic T-lymphocyte antigen-4 (CTLA4), by treating patients with Pembrolizumab to enhance the immune response towards tumor cells (186, 244, 245). The KEYNOTE-028 phase I study treated PDL-1 positive PNEN patients with Pembrolizumab and reported an ORR of 6.3%, but no CRs occurred. Responses were better in metastatic carcinoids (ORR: 12%) (246). These findings were confirmed by the KEYNOTE-158 phase II study, in which the ORR was 3.7% with 3 PRs in PNEN and 1 in rectal NEN patients (247). Currently, several other clinical trials that are investigating the antitumor effect of immune checkpoint inhibitors include NCT02939651 for Pembrolizumab and NCT02955069 for other PDL-1 receptor antibodies (247). Moreover, there is also much speculation that PRRT cytotoxic drugs induce genotoxicity, hence increase the neoantigen load and thereby could potentially enhance the efficacy of immunotherapy (149, 244, 245, 248). Bevacizumab, a monoclonal antibody against the VEGF, showed no real benefit in PFS in a phase III trial in which Bevacizumab + Octreotide was compared to Interferon + Octreotide (249). The BETTER phase II trial, on the other hand, demonstrated that Bevacizumab + 5-FU/Streptozocin in patients with metastatic, well-differentiated PNENs could reach a PFS of 23.7 months and they reported an OS at 24 months of 88% (250). The SANET-ep (251) and SANET-p (252) phase III studies examined the efficacy and safety of Surufatinib in extrapancreatic NENs and PNENs, respectively. Surufatinib, a small-molecule inhibitor that targets VEGFRs as well as the fibroblast growth factor (FGF) receptor 1 and macrophage colony-stimulating factor 1 (CSF1) receptor, effectively prolonged PFS in both studies and was therefore suggested a potential treatment option in both patient populations (251, 252).

 

QUALITY OF LIFE IN PATIENTS WITH PNENs

 

The measurement of health-related quality of life (HRQOL) has become essential for evaluating the impact of the disease process and the treatment on patient’s symptoms, social, emotional, physiological, and physical functioning. The European Organization for Research and Treatment of Cancer (EORTC) developed the QLQ-C30 tool for oncology patients (253) and the QLQ-GINET21 tool was specifically developed for a spectrum of NEN patients (28% PNENs) (254). The Norfolk QOLNET was specifically developed for midgut NETs (carcinoids) and may provide some additional advantages for that specific group of patients (231).

 

The most commonly used QOL tool in GEP-NENs (including PNENs) is the EORTC QLQ-C30 (255). SSAs and Sunitinib have shown to improve HRQOL in diverse groups of GEP-NEN patients (255). In the CLARINET study, QLC-C30 data were mapped to EQ-5D utilities and not surprisingly worse utility values were noted with PD compared to SD. Of note, tumor location (midgut vs. pancreas) did not affect utility (256). PNEN patients treated with everolimus showed stable HRQOL scores as opposed to worse scores in non-PNEN patients (Pavel). PRRT treatment of PNEN patients resulted in significantly improved global health status, social functioning and mitigation of physical complaints (257). Thus, data are emerging on HRQOL in PNEN patients. However, most studies are too heterogenous in terms of patient populations and treatment interventions to draw firm conclusions (258). Moving forward, it will be important for HRQOL to be measured as a key component of clinical trials.

 

EXPERT COMMENTARY  

 

After many years of frustration, our knowledge regarding the biology, pathophysiology and genetics of (P)NENs increased. This has led to marked improvements in (functional) imaging, with the development of 68Gallium-labeled SSAs, as well as targeted treatments such as the tyrosine-kinase inhibitor Sunitinib, and the mTOR inhibitor Everolimus. In addition, PRRT seems to be expanding its role in treatment from midgut to PNENs. In the future, both imaging and treatment options will continue to evolve as more specific imaging agents and therapeutic targets are being developed and evaluated in numerous studies. The relatively uncommon nature of PNENs has made designing and completing randomized studies of adequate power challenging for a single institution. Therefore, we encourage the recent trend of multi-institutional, multinational studies in more homogeneous patient populations. We also strongly agree with the recommendation of NANETS, ENETS and other groups that all of these patients should be entered into clinical trials whenever feasible. Determining study availability and patient eligibility has been greatly facilitated by Clinical trials.gov as well as institutional and organizational websites. Enrolling more patients in clinical trials by overcoming barriers to participation will be required to move patient care forward.

 

Unfortunately, to date, the optimal treatment(s) and treatment sequences have yet to be defined. The lack of treatment standardization, the plethora of treatments that most patients receive, and different treatment sequences make it extremely difficult to assess the effectiveness of a particular treatment relative to others. Moreover, head-to-head comparisons are lacking as well. Available consensus guidelines establish broad principles, but are generally not helpful in managing a specific patient. Management has become even more complex given the multiplicity of effective treatments for advanced disease, none of which has convincingly been shown to be superior to the other. Hence, an experienced multidisciplinary team is essential to guide management of these patients. Given relative parity of effectiveness, decisions regarding choice of treatment need to be based on multiple considerations, including patient’s overall health, disease burden, symptomatology, rate of progression, treatment toxicity, effect on QOL, and cost. These considerations will usually lead to one treatment being favored over another.

 

Because of the relatively indolent nature of many (or most) NENs, long-term follow-up to assess differences in treatment outcomes, is required. However, biomarkers that can predict response to a particular therapy are currently not available. We expect that in the future the so-called tumor circulome, especially ctDNA, could play an important role in this as recent studies revealed its potential to diagnose, prognosticate and monitor disease progression.

 

5 Year View

 

Knowledge of the biology and genetics will continue to accumulate, which could potentially lead to further refinements in classification, staging, and personalized treatment. Genetic profiling will become clinically useful as data will accumulate on treatment effectiveness in patient subgroups leading to more tailored therapies. Moreover, biomarkers will be developed that better predict response to a particular therapy. Results of the ongoing clinical trials on newer SSAs and targeted agents will add to the number of available treatments. The role of PRRT in the treatment of PNENs will be better defined. There will be increased knowledge as to optimal treatment sequences. Designing randomized clinical trials of adequate power will remain a challenge for many reasons including the scarcity and indolent growth of these tumors. Consensus guidelines will evolve, but patient management will continue to require an experienced multidisciplinary team.

 

REFERENCES

 

  1. Clift AK, Kidd M, Bodei L, Toumpanakis C, Baum RP, Oberg K, et al. Neuroendocrine Neoplasms of the Small Bowel and Pancreas. Neuroendocrinology. 2020;110(6):444-76.
  2. Metz DC, Jensen RT. Gastrointestinal neuroendocrine tumors: pancreatic endocrine tumors. Gastroenterology. 2008;135(5):1469-92.
  3. Perri G, Prakash LR, Katz MHG. Pancreatic neuroendocrine tumors. Current opinion in gastroenterology. 2019;35(5):468-77.
  4. Hong X, Qiao S, Li F, Wang W, Jiang R, Wu H, et al. Whole-genome sequencing reveals distinct genetic bases for insulinomas and non-functional pancreatic neuroendocrine tumours: leading to a new classification system. Gut. 2020;69(5):877-87.
  5. Dasari A, Shen C, Halperin D, Zhao B, Zhou S, Xu Y, et al. Trends in the Incidence, Prevalence, and Survival Outcomes in Patients With Neuroendocrine Tumors in the United States. JAMA oncology. 2017;3(10):1335-42.
  6. Daskalakis K. Functioning and nonfunctioning pNENs. Current Opinion in Endocrine and Metabolic Research. 2021;18:284-90.
  7. Ma ZY, Gong YF, Zhuang HK, Zhou ZX, Huang SZ, Zou YP, et al. Pancreatic neuroendocrine tumors: A review of serum biomarkers, staging, and management. World journal of gastroenterology. 2020;26(19):2305-22.
  8. Cives M, Strosberg JR. Gastroenteropancreatic Neuroendocrine Tumors. CA: a cancer journal for clinicians. 2018;68(6):471-87.
  9. Falconi M, Eriksson B, Kaltsas G, Bartsch DK, Capdevila J, Caplin M, et al. ENETS Consensus Guidelines Update for the Management of Patients with Functional Pancreatic Neuroendocrine Tumors and Non-Functional Pancreatic Neuroendocrine Tumors. Neuroendocrinology. 2016;103(2):153-71.
  10. Boons G, Vandamme T, Peeters M, Van Camp G, Op de Beeck K. Clinical applications of (epi)genetics in gastroenteropancreatic neuroendocrine neoplasms: Moving towards liquid biopsies. Reviews in endocrine & metabolic disorders. 2019;20(3):333-51.
  11. Liakakos T, Roukos DH. Everolimus and sunitinib: from mouse models to treatment of pancreatic neuroendocrine tumors. Future oncology (London, England). 2011;7(9):1025-9.
  12. Vinik AI. Advances in diagnosis and treatment of pancreatic neuroendocrine tumors. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists. 2014;20(11):1222-30.
  13. Verbeke CS. Endocrine tumours of the pancreas. Histopathology. 2010;56(6):669-82.
  14. Plöckinger U, Wiedenmann B. Diagnosis of non-functioning neuro-endocrine gastro-enteropancreatic tumours. Neuroendocrinology. 2004;80 Suppl 1:35-8.
  15. Inzani F, Petrone G, Rindi G. The New World Health Organization Classification for Pancreatic Neuroendocrine Neoplasia. Endocrinology and metabolism clinics of North America. 2018;47(3):463-70.
  16. Nagtegaal ID, Odze RD, Klimstra D, Paradis V, Rugge M, Schirmacher P, et al. The 2019 WHO classification of tumours of the digestive system. Histopathology. 2020;76(2):182-8.
  17. Ciobanu OA, Martin S, Fica S. Perspectives on the diagnostic, predictive and prognostic markers of neuroendocrine neoplasms (Review). Experimental and therapeutic medicine. 2021;22(6):1479.
  18. Jensen RT, Berna MJ, Bingham DB, Norton JA. Inherited pancreatic endocrine tumor syndromes: advances in molecular pathogenesis, diagnosis, management, and controversies. Cancer. 2008;113(7 Suppl):1807-43.
  19. Li X, Gou S, Liu Z, Ye Z, Wang C. Assessment of the American Joint Commission on Cancer 8th Edition Staging System for Patients with Pancreatic Neuroendocrine Tumors: A Surveillance, Epidemiology, and End Results analysis. Cancer medicine. 2018;7(3):626-34.
  20. You Y, Jang JY, Kim SC, Yoon YS, Park JS, Cho CK, et al. Validation of the 8th AJCC Cancer Staging System for Pancreas Neuroendocrine Tumors Using Korean Nationwide Surgery Database. Cancer research and treatment. 2019;51(4):1639-52.
  21. Amin MB, Greene FL, Edge SB, Compton CC, Gershenwald JE, Brookland RK, et al. The Eighth Edition AJCC Cancer Staging Manual: Continuing to build a bridge from a population-based to a more "personalized" approach to cancer staging. CA: a cancer journal for clinicians. 2017;67(2):93-9.
  22. Yang M, Zeng L, Yao WQ, Ke NW, Tan CL, Tian BL, et al. A comprehensive validation of the novel 8th edition of American Joint Committee on Cancer staging manual for the long-term survivals of patients with non-functional pancreatic neuroendocrine neoplasms. Medicine. 2020;99(46):e22291.
  23. Reid MD, Balci S, Saka B, Adsay NV. Neuroendocrine tumors of the pancreas: current concepts and controversies. Endocrine pathology. 2014;25(1):65-79.
  24. Di Domenico A, Pipinikas CP, Maire RS, Bräutigam K, Simillion C, Dettmer MS, et al. Epigenetic landscape of pancreatic neuroendocrine tumours reveals distinct cells of origin and means of tumour progression. Communications biology. 2020;3(1):740.
  25. Noone TC, Hosey J, Firat Z, Semelka RC. Imaging and localization of islet-cell tumours of the pancreas on CT and MRI. Best practice & research Clinical endocrinology & metabolism. 2005;19(2):195-211.
  26. Taskin OC, Clarke CN, Erkan M, Tsai S, Evans DB, Adsay V. Pancreatic neuroendocrine neoplasms: current state and ongoing controversies on terminology, classification and prognostication. Journal of gastrointestinal oncology. 2020;11(3):548-58.
  27. Gao HL, Wang WQ, Yu XJ, Liu L. Molecular drivers and cells of origin in pancreatic ductal adenocarcinoma and pancreatic neuroendocrine carcinoma. Experimental hematology & oncology. 2020;9:28.
  28. Chan CS, Laddha SV, Lewis PW, Koletsky MS, Robzyk K, Da Silva E, et al. ATRX, DAXX or MEN1 mutant pancreatic neuroendocrine tumors are a distinct alpha-cell signature subgroup. Nature communications. 2018;9(1):4158.
  29. Cejas P, Drier Y, Dreijerink KMA, Brosens LAA, Deshpande V, Epstein CB, et al. Enhancer signatures stratify and predict outcomes of non-functional pancreatic neuroendocrine tumors. Nature medicine. 2019;25(8):1260-5.
  30. Strobel O, Rosow DE, Rakhlin EY, Lauwers GY, Trainor AG, Alsina J, et al. Pancreatic duct glands are distinct ductal compartments that react to chronic injury and mediate Shh-induced metaplasia. Gastroenterology. 2010;138(3):1166-77.
  31. Anderson CW, Bennett JJ. Clinical Presentation and Diagnosis of Pancreatic Neuroendocrine Tumors. Surgical oncology clinics of North America. 2016;25(2):363-74.
  32. Mimeault M, Batra SK. Recent progress on normal and malignant pancreatic stem/progenitor cell research: therapeutic implications for the treatment of type 1 or 2 diabetes mellitus and aggressive pancreatic cancer. Gut. 2008;57(10):1456-68.
  33. Jiang FX, Morahan G. Multipotent pancreas progenitors: Inconclusive but pivotal topic. World journal of stem cells. 2015;7(11):1251-61.
  34. Zhong F, Jiang Y. Endogenous Pancreatic β Cell Regeneration: A Potential Strategy for the Recovery of β Cell Deficiency in Diabetes. Frontiers in endocrinology. 2019;10:101.
  35. Yamaguchi J, Liss AS, Sontheimer A, Mino-Kenudson M, Castillo CF, Warshaw AL, et al. Pancreatic duct glands (PDGs) are a progenitor compartment responsible for pancreatic ductal epithelial repair. Stem cell research. 2015;15(1):190-202.
  36. Tirosh A, Kebebew E. Genetic and epigenetic alterations in pancreatic neuroendocrine tumors. Journal of gastrointestinal oncology. 2020;11(3):567-77.
  37. Görtz B, Roth J, Krähenmann A, de Krijger RR, Muletta-Feurer S, Rütimann K, et al. Mutations and allelic deletions of the MEN1 gene are associated with a subset of sporadic endocrine pancreatic and neuroendocrine tumors and not restricted to foregut neoplasms. The American journal of pathology. 1999;154(2):429-36.
  38. Mafficini A, Scarpa A. Genetics and Epigenetics of Gastroenteropancreatic Neuroendocrine Neoplasms. Endocrine reviews. 2019;40(2):506-36.
  39. Scarpa A, Chang DK, Nones K, Corbo V, Patch AM, Bailey P, et al. Whole-genome landscape of pancreatic neuroendocrine tumours. Nature. 2017;543(7643):65-71.
  40. Crona J, Skogseid B. GEP- NETS UPDATE: Genetics of neuroendocrine tumors. European journal of endocrinology. 2016;174(6):R275-90.
  41. Jiao Y, Shi C, Edil BH, de Wilde RF, Klimstra DS, Maitra A, et al. DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science (New York, NY). 2011;331(6021):1199-203.
  42. Gläsker S, Vergauwen E, Koch CA, Kutikov A, Vortmeyer AO. Von Hippel-Lindau Disease: Current Challenges and Future Prospects. OncoTargets and therapy. 2020;13:5669-90.
  43. Zwolak A, Świrska J, Tywanek E, Dudzińska M, Tarach JS, Matyjaszek-Matuszek B. Pancreatic neuroendocrine tumours in patients with von Hippel-Lindau disease. Endokrynologia Polska. 2020;71(3):256-9.
  44. Parekh VI, Modali SD, Welch J, Simonds WF, Weinstein LS, Kebebew E, et al. Frequency and consequence of the recurrent YY1 p.T372R mutation in sporadic insulinomas. Endocrine-related cancer. 2018;25(5):L31-l5.
  45. Pipinikas CP, Berner AM, Sposito T, Thirlwell C. The evolving (epi)genetic landscape of pancreatic neuroendocrine tumours. Endocrine-related cancer. 2019;26(9):R519-r44.
  46. Singhi AD, Klimstra DS. Well-differentiated pancreatic neuroendocrine tumours (PanNETs) and poorly differentiated pancreatic neuroendocrine carcinomas (PanNECs): concepts, issues and a practical diagnostic approach to high-grade (G3) cases. Histopathology. 2018;72(1):168-77.
  47. Boons G, Vandamme T, Mariën L, Lybaert W, Roeyen G, Rondou T, et al. Longitudinal Copy-Number Alteration Analysis in Plasma Cell-Free DNA of Neuroendocrine Neoplasms is a Novel Specific Biomarker for Diagnosis, Prognosis, and Follow-up. Clinical cancer research : an official journal of the American Association for Cancer Research. 2021.
  48. House MG, Herman JG, Guo MZ, Hooker CM, Schulick RD, Lillemoe KD, et al. Aberrant hypermethylation of tumor suppressor genes in pancreatic endocrine neoplasms. Annals of surgery. 2003;238(3):423-31; discussion 31-2.
  49. Neiman D, Moss J, Hecht M, Magenheim J, Piyanzin S, Shapiro AMJ, et al. Islet cells share promoter hypomethylation independently of expression, but exhibit cell-type-specific methylation in enhancers. Proceedings of the National Academy of Sciences of the United States of America. 2017;114(51):13525-30.
  50. Boons G, Vandamme T, Ibrahim J, Roeyen G, Driessen A, Peeters D, et al. PDX1 DNA Methylation Distinguishes Two Subtypes of Pancreatic Neuroendocrine Neoplasms with a Different Prognosis. Cancers. 2020;12(6).
  51. Lakis V, Lawlor RT, Newell F, Patch AM, Mafficini A, Sadanandam A, et al. DNA methylation patterns identify subgroups of pancreatic neuroendocrine tumors with clinical association. Communications biology. 2021;4(1):155.
  52. Scott AT, Howe JR. Evaluation and Management of Neuroendocrine Tumors of the Pancreas. The Surgical clinics of North America. 2019;99(4):793-814.
  53. Lee DW, Kim MK, Kim HG. Diagnosis of Pancreatic Neuroendocrine Tumors. Clinical endoscopy. 2017;50(6):537-45.
  54. Rebours V, Cordova J, Couvelard A, Fabre M, Palazzo L, Vullierme MP, et al. Can pancreatic neuroendocrine tumour biopsy accurately determine pathological characteristics? Digestive and liver disease : official journal of the Italian Society of Gastroenterology and the Italian Association for the Study of the Liver. 2015;47(11):973-7.
  55. Klöppel G, Couvelard A, Perren A, Komminoth P, McNicol AM, Nilsson O, et al. ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Tumors: towards a standardized approach to the diagnosis of gastroenteropancreatic neuroendocrine tumors and their prognostic stratification. Neuroendocrinology. 2009;90(2):162-6.
  56. Eusebi LH, Thorburn D, Toumpanakis C, Frazzoni L, Johnson G, Vessal S, et al. Endoscopic ultrasound-guided fine-needle aspiration vs fine-needle biopsy for the diagnosis of pancreatic neuroendocrine tumors. Endoscopy international open. 2019;7(11):E1393-e9.
  57. Crinò SF, Ammendola S, Meneghetti A, Bernardoni L, Conti Bellocchi MC, Gabbrielli A, et al. Comparison between EUS-guided fine-needle aspiration cytology and EUS-guided fine-needle biopsy histology for the evaluation of pancreatic neuroendocrine tumors. Pancreatology : official journal of the International Association of Pancreatology (IAP)[et al]. 2021;21(2):443-50.
  58. Garcia-Carbonero R, Sorbye H, Baudin E, Raymond E, Wiedenmann B, Niederle B, et al. ENETS Consensus Guidelines for High-Grade Gastroenteropancreatic Neuroendocrine Tumors and Neuroendocrine Carcinomas. Neuroendocrinology. 2016;103(2):186-94.
  59. Fang JM, Shi J. A Clinicopathologic and Molecular Update of Pancreatic Neuroendocrine Neoplasms With a Focus on the New World Health Organization Classification. Archives of pathology & laboratory medicine. 2019;143(11):1317-26.
  60. Perren A, Couvelard A, Scoazec JY, Costa F, Borbath I, Delle Fave G, et al. ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Tumors: Pathology: Diagnosis and Prognostic Stratification. Neuroendocrinology. 2017;105(3):196-200.
  61. Rockall AG, Reznek RH. Imaging of neuroendocrine tumours (CT/MR/US). Best practice & research Clinical endocrinology & metabolism. 2007;21(1):43-68.
  62. Virgolini I, Traub-Weidinger T, Decristoforo C. Nuclear medicine in the detection and management of pancreatic islet-cell tumours. Best practice & research Clinical endocrinology & metabolism. 2005;19(2):213-27.
  63. Gibril F, Jensen RT. Diagnostic uses of radiolabelled somatostatin receptor analogues in gastroenteropancreatic endocrine tumours. Digestive and liver disease : official journal of the Italian Society of Gastroenterology and the Italian Association for the Study of the Liver. 2004;36 Suppl 1:S106-20.
  64. Sundin A, Garske U, Orlefors H. Nuclear imaging of neuroendocrine tumours. Best practice & research Clinical endocrinology & metabolism. 2007;21(1):69-85.
  65. Singh A, Hines JJ, Friedman B. Multimodality Imaging of the Pancreatic Neuroendocrine Tumors. Seminars in Ultrasound, CT and MRI. 2019;40(6):469-82.
  66. Chiti G, Grazzini G, Cozzi D, Danti G, Matteuzzi B, Granata V, et al. Imaging of Pancreatic Neuroendocrine Neoplasms. International journal of environmental research and public health. 2021;18(17).
  67. Morse B, Al-Toubah T, Montilla-Soler J. Anatomic and Functional Imaging of Neuroendocrine Tumors. Current treatment options in oncology. 2020;21(9):75.
  68. Khanna L, Prasad SR, Sunnapwar A, Kondapaneni S, Dasyam A, Tammisetti VS, et al. Pancreatic Neuroendocrine Neoplasms: 2020 Update on Pathologic and Imaging Findings and Classification. Radiographics : a review publication of the Radiological Society of North America, Inc. 2020;40(5):1240-62.
  69. Rozenblum L, Mokrane FZ, Yeh R, Sinigaglia M, Besson FL, Seban RD, et al. Imaging-guided precision medicine in non-resectable gastro-entero-pancreatic neuroendocrine tumors: A step-by-step approach. European journal of radiology. 2020;122:108743.
  70. Galgano SJ, Sharbidre K, Morgan DE. Multimodality Imaging of Neuroendocrine Tumors. Radiologic clinics of North America. 2020;58(6):1147-59.
  71. Tamm EP, Bhosale P, Lee JH, Rohren EM. State-of-the-art Imaging of Pancreatic Neuroendocrine Tumors. Surgical oncology clinics of North America. 2016;25(2):375-400.
  72. Sundin A, Vullierme MP, Kaltsas G, Plöckinger U. ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Tumors: radiological examinations. Neuroendocrinology. 2009;90(2):167-83.
  73. Jackson JE. Angiography and arterial stimulation venous sampling in the localization of pancreatic neuroendocrine tumours. Best practice & research Clinical endocrinology & metabolism. 2005;19(2):229-39.
  74. McLean AM, Fairclough PD. Endoscopic ultrasound in the localisation of pancreatic islet cell tumours. Best practice & research Clinical endocrinology & metabolism. 2005;19(2):177-93.
  75. Norton JA, Jensen RT. Resolved and unresolved controversies in the surgical management of patients with Zollinger-Ellison syndrome. Annals of surgery. 2004;240(5):757-73.
  76. Krenning EP, Kwekkeboom DJ, Oei HY, de Jong RJ, Dop FJ, Reubi JC, et al. Somatostatin-receptor scintigraphy in gastroenteropancreatic tumors. An overview of European results. Annals of the New York Academy of Sciences. 1994;733:416-24.
  77. Desai H, Borges-Neto S, Wong TZ. Molecular Imaging and Therapy for Neuroendocrine Tumors. Current treatment options in oncology. 2019;20(10):78.
  78. Termanini B, Gibril F, Reynolds JC, Doppman JL, Chen CC, Stewart CA, et al. Value of somatostatin receptor scintigraphy: a prospective study in gastrinoma of its effect on clinical management. Gastroenterology. 1997;112(2):335-47.
  79. Gibril F, Reynolds JC, Chen CC, Yu F, Goebel SU, Serrano J, et al. Specificity of somatostatin receptor scintigraphy: a prospective study and effects of false-positive localizations on management in patients with gastrinomas. Journal of nuclear medicine : official publication, Society of Nuclear Medicine. 1999;40(4):539-53.
  80. Christ E, Wild D, Ederer S, Béhé M, Nicolas G, Caplin ME, et al. Glucagon-like peptide-1 receptor imaging for the localisation of insulinomas: a prospective multicentre imaging study. The lancet Diabetes & endocrinology. 2013;1(2):115-22.
  81. Panagiotidis E, Alshammari A, Michopoulou S, Skoura E, Naik K, Maragkoudakis E, et al. Comparison of the Impact of 68Ga-DOTATATE and 18F-FDG PET/CT on Clinical Management in Patients with Neuroendocrine Tumors. Journal of nuclear medicine : official publication, Society of Nuclear Medicine. 2017;58(1):91-6.
  82. Maxwell JE, Howe JR. Imaging in neuroendocrine tumors: an update for the clinician. International journal of endocrine oncology. 2015;2(2):159-68.
  83. Johnbeck CB, Knigge U, Kjær A. PET tracers for somatostatin receptor imaging of neuroendocrine tumors: current status and review of the literature. Future oncology (London, England). 2014;10(14):2259-77.
  84. Kornaczewski Jackson ER, Pointon OP, Bohmer R, Burgess JR. Utility of FDG-PET Imaging for Risk Stratification of Pancreatic Neuroendocrine Tumors in MEN1. The Journal of clinical endocrinology and metabolism. 2017;102(6):1926-33.
  85. Orlefors H, Sundin A, Garske U, Juhlin C, Oberg K, Skogseid B, et al. Whole-body (11)C-5-hydroxytryptophan positron emission tomography as a universal imaging technique for neuroendocrine tumors: comparison with somatostatin receptor scintigraphy and computed tomography. The Journal of clinical endocrinology and metabolism. 2005;90(6):3392-400.
  86. Gabriel M, Decristoforo C, Kendler D, Dobrozemsky G, Heute D, Uprimny C, et al. 68Ga-DOTA-Tyr3-octreotide PET in neuroendocrine tumors: comparison with somatostatin receptor scintigraphy and CT. Journal of nuclear medicine : official publication, Society of Nuclear Medicine. 2007;48(4):508-18.
  87. Eriksson B, Orlefors H, Oberg K, Sundin A, Bergström M, Långström B. Developments in PET for the detection of endocrine tumours. Best practice & research Clinical endocrinology & metabolism. 2005;19(2):311-24.
  88. Geijer H, Breimer LH. Somatostatin receptor PET/CT in neuroendocrine tumours: update on systematic review and meta-analysis. European journal of nuclear medicine and molecular imaging. 2013;40(11):1770-80.
  89. Deppen SA, Liu E, Blume JD, Clanton J, Shi C, Jones-Jackson LB, et al. Safety and Efficacy of 68Ga-DOTATATE PET/CT for Diagnosis, Staging, and Treatment Management of Neuroendocrine Tumors. Journal of nuclear medicine : official publication, Society of Nuclear Medicine. 2016;57(5):708-14.
  90. Sadowski SM, Neychev V, Millo C, Shih J, Nilubol N, Herscovitch P, et al. Prospective Study of 68Ga-DOTATATE Positron Emission Tomography/Computed Tomography for Detecting Gastro-Entero-Pancreatic Neuroendocrine Tumors and Unknown Primary Sites. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2016;34(6):588-96.
  91. Etchebehere EC, de Oliveira Santos A, Gumz B, Vicente A, Hoff PG, Corradi G, et al. 68Ga-DOTATATE PET/CT, 99mTc-HYNIC-octreotide SPECT/CT, and whole-body MR imaging in detection of neuroendocrine tumors: a prospective trial. Journal of nuclear medicine : official publication, Society of Nuclear Medicine. 2014;55(10):1598-604.
  92. Skoura E, Michopoulou S, Mohmaduvesh M, Panagiotidis E, Al Harbi M, Toumpanakis C, et al. The Impact of 68Ga-DOTATATE PET/CT Imaging on Management of Patients with Neuroendocrine Tumors: Experience from a National Referral Center in the United Kingdom. Journal of nuclear medicine : official publication, Society of Nuclear Medicine. 2016;57(1):34-40.
  93. Crown A, Rocha FG, Raghu P, Lin B, Funk G, Alseidi A, et al. Impact of initial imaging with gallium-68 dotatate PET/CT on diagnosis and management of patients with neuroendocrine tumors. Journal of surgical oncology. 2020;121(3):480-5.
  94. Hofman MS, Lau WF, Hicks RJ. Somatostatin receptor imaging with 68Ga DOTATATE PET/CT: clinical utility, normal patterns, pearls, and pitfalls in interpretation. Radiographics : a review publication of the Radiological Society of North America, Inc. 2015;35(2):500-16.
  95. Lastoria S, Marciello F, Faggiano A, Aloj L, Caracò C, Aurilio M, et al. Role of (68)Ga-DOTATATE PET/CT in patients with multiple endocrine neoplasia type 1 (MEN1). Endocrine. 2016;52(3):488-94.
  96. Luo Y, Pan Q, Yao S, Yu M, Wu W, Xue H, et al. Glucagon-Like Peptide-1 Receptor PET/CT with 68Ga-NOTA-Exendin-4 for Detecting Localized Insulinoma: A Prospective Cohort Study. Journal of nuclear medicine : official publication, Society of Nuclear Medicine. 2016;57(5):715-20.
  97. Elazezy M, Joosse SA. Techniques of using circulating tumor DNA as a liquid biopsy component in cancer management. Computational and structural biotechnology journal. 2018;16:370-8.
  98. Siravegna G, Marsoni S, Siena S, Bardelli A. Integrating liquid biopsies into the management of cancer. Nature reviews Clinical oncology. 2017;14(9):531-48.
  99. De Rubis G, Rajeev Krishnan S, Bebawy M. Liquid Biopsies in Cancer Diagnosis, Monitoring, and Prognosis. Trends in pharmacological sciences. 2019;40(3):172-86.
  100. Heitzer E, Haque IS, Roberts CES, Speicher MR. Current and future perspectives of liquid biopsies in genomics-driven oncology. Nature reviews Genetics. 2019;20(2):71-88.
  101. Lissa D, Robles AI. Methylation analyses in liquid biopsy. Translational lung cancer research. 2016;5(5):492-504.
  102. Bocchini M, Nicolini F, Severi S, Bongiovanni A, Ibrahim T, Simonetti G, et al. Biomarkers for Pancreatic Neuroendocrine Neoplasms (PanNENs) Management-An Updated Review. Frontiers in oncology. 2020;10:831.
  103. Öberg K, Califano A, Strosberg JR, Ma S, Pape U, Bodei L, et al. A meta-analysis of the accuracy of a neuroendocrine tumor mRNA genomic biomarker (NETest) in blood. Annals of oncology : official journal of the European Society for Medical Oncology. 2020;31(2):202-12.
  104. Oberg K, Modlin IM, De Herder W, Pavel M, Klimstra D, Frilling A, et al. Consensus on biomarkers for neuroendocrine tumour disease. The Lancet Oncology. 2015;16(9):e435-e46.
  105. Parbhu SK, Adler DG. Pancreatic neuroendocrine tumors: contemporary diagnosis and management. Hospital practice (1995). 2016;44(3):109-19.
  106. de Herder WW. Biochemistry of neuroendocrine tumours. Best practice & research Clinical endocrinology & metabolism. 2007;21(1):33-41.
  107. Ardill JE. Circulating markers for endocrine tumours of the gastroenteropancreatic tract. Annals of clinical biochemistry. 2008;45(Pt 6):539-59.
  108. Oberg K. Biochemical diagnosis of neuroendocrine GEP tumor. The Yale journal of biology and medicine. 1997;70(5-6):501-8.
  109. Panzuto F, Severi C, Cannizzaro R, Falconi M, Angeletti S, Pasquali A, et al. Utility of combined use of plasma levels of chromogranin A and pancreatic polypeptide in the diagnosis of gastrointestinal and pancreatic endocrine tumors. Journal of endocrinological investigation. 2004;27(1):6-11.
  110. Bajetta E, Ferrari L, Martinetti A, Celio L, Procopio G, Artale S, et al. Chromogranin A, neuron specific enolase, carcinoembryonic antigen, and hydroxyindole acetic acid evaluation in patients with neuroendocrine tumors. Cancer. 1999;86(5):858-65.
  111. Stabile BE, Howard TJ, Passaro E, Jr., O'Connor DT. Source of plasma chromogranin A elevation in gastrinoma patients. Archives of surgery (Chicago, Ill : 1960). 1990;125(4):451-3.
  112. Qiao XW, Qiu L, Chen YJ, Meng CT, Sun Z, Bai CM, et al. Chromogranin A is a reliable serum diagnostic biomarker for pancreatic neuroendocrine tumors but not for insulinomas. BMC endocrine disorders. 2014;14:64.
  113. Nobels FR, Kwekkeboom DJ, Coopmans W, Schoenmakers CH, Lindemans J, De Herder WW, et al. Chromogranin A as serum marker for neuroendocrine neoplasia: comparison with neuron-specific enolase and the alpha-subunit of glycoprotein hormones. The Journal of clinical endocrinology and metabolism. 1997;82(8):2622-8.
  114. Bílek R, Šafařík L, Ciprová V, Vlcek P, Lisá L. Chromogranin A, a member of neuroendocrine secretory proteins as a selective marker for laboratory diagnosis of pheochromocytoma. Physiological research. 2008;57 Suppl 1:S171-s9.
  115. Marotta V, Zatelli MC, Sciammarella C, Ambrosio MR, Bondanelli M, Colao A, et al. Chromogranin A as circulating marker for diagnosis and management of neuroendocrine neoplasms: more flaws than fame. Endocrine-related cancer. 2018;25(1):R11-r29.
  116. Stridsberg M, Eriksson B, Fellström B, Kristiansson G, Tiensuu Janson E. Measurements of chromogranin B can serve as a complement to chromogranin A. Regulatory peptides. 2007;139(1-3):80-3.
  117. Takiyyuddin MA, Cervenka JH, Hsiao RJ, Barbosa JA, Parmer RJ, O'Connor DT. Chromogranin A. Storage and release in hypertension. Hypertension (Dallas, Tex : 1979). 1990;15(3):237-46.
  118. Lindholm DP, Oberg K. Biomarkers and molecular imaging in gastroenteropancreatic neuroendocrine tumors. Hormone and metabolic research = Hormon- und Stoffwechselforschung = Hormones et metabolisme. 2011;43(12):832-7.
  119. Jilesen AP, Busch OR, van Gulik TM, Gouma DJ, Nieveen van Dijkum EJ. Standard pre- and postoperative determination of chromogranin a in resectable non-functioning pancreatic neuroendocrine tumors--diagnostic accuracy: NF-pNET and low tumor burden. Digestive surgery. 2014;31(6):407-14.
  120. Monaghan PJ, Lamarca A, Valle JW, Hubner RA, Mansoor W, Trainer PJ, et al. Routine measurement of plasma chromogranin B has limited clinical utility in the management of patients with neuroendocrine tumours. Clinical endocrinology. 2016;84(3):348-52.
  121. Hofland J, Zandee WT, de Herder WW. Role of biomarker tests for diagnosis of neuroendocrine tumours. Nature reviews Endocrinology. 2018;14(11):656-69.
  122. van Adrichem RC, Kamp K, Vandamme T, Peeters M, Feelders RA, de Herder WW. Serum neuron-specific enolase level is an independent predictor of overall survival in patients with gastroenteropancreatic neuroendocrine tumors. Annals of oncology : official journal of the European Society for Medical Oncology. 2016;27(4):746-7.
  123. Stronge RL, Turner GB, Johnston BT, McCance DR, McGinty A, Patterson CC, et al. A rapid rise in circulating pancreastatin in response to somatostatin analogue therapy is associated with poor survival in patients with neuroendocrine tumours. Annals of clinical biochemistry. 2008;45(Pt 6):560-6.
  124. Walter T, Chardon L, Chopin-laly X, Raverot V, Caffin AG, Chayvialle JA, et al. Is the combination of chromogranin A and pancreatic polypeptide serum determinations of interest in the diagnosis and follow-up of gastro-entero-pancreatic neuroendocrine tumours? European journal of cancer (Oxford, England : 1990). 2012;48(12):1766-73.
  125. Brown JE, Cook RJ, Major P, Lipton A, Saad F, Smith M, et al. Bone turnover markers as predictors of skeletal complications in prostate cancer, lung cancer, and other solid tumors. Journal of the National Cancer Institute. 2005;97(1):59-69.
  126. Lipton A, Costa L, Ali S, Demers L. Use of markers of bone turnover for monitoring bone metastases and the response to therapy. Seminars in oncology. 2001;28(4 Suppl 11):54-9.
  127. Khan MS, Kirkwood A, Tsigani T, Garcia-Hernandez J, Hartley JA, Caplin ME, et al. Circulating tumor cells as prognostic markers in neuroendocrine tumors. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2013;31(3):365-72.
  128. Mandair D, Khan MS, Lopes A, Furtado O'Mahony L, Ensell L, Lowe H, et al. Prognostic Threshold for Circulating Tumor Cells in Patients With Pancreatic and Midgut Neuroendocrine Tumors. The Journal of clinical endocrinology and metabolism. 2021;106(3):872-82.
  129. Khan MS, Tsigani T, Rashid M, Rabouhans JS, Yu D, Luong TV, et al. Circulating tumor cells and EpCAM expression in neuroendocrine tumors. Clinical cancer research : an official journal of the American Association for Cancer Research. 2011;17(2):337-45.
  130. Rizzo FM, Vesely C, Childs A, Marafioti T, Khan MS, Mandair D, et al. Circulating tumour cells and their association with bone metastases in patients with neuroendocrine tumours. British journal of cancer. 2019;120(3):294-300.
  131. Grande E, Capdevila J, Castellano D, Teulé A, Durán I, Fuster J, et al. Pazopanib in pretreated advanced neuroendocrine tumors: a phase II, open-label trial of the Spanish Task Force Group for Neuroendocrine Tumors (GETNE). Annals of oncology : official journal of the European Society for Medical Oncology. 2015;26(9):1987-93.
  132. Meyer T, Caplin M, Khan MS, Toumpanakis C, Shetty S, Ramage JK, et al. Circulating tumor cells and tumor biomarkers in functional midgut neuroendocrine tumors. Journal of Neuroendocrinology. 2022.
  133. Childs A, Vesely C, Ensell L, Lowe H, Luong TV, Caplin ME, et al. Expression of somatostatin receptors 2 and 5 in circulating tumour cells from patients with neuroendocrine tumours. British journal of cancer. 2016;115(12):1540-7.
  134. Rizzo FM, Meyer T. Liquid Biopsies for Neuroendocrine Tumors: Circulating Tumor Cells, DNA, and MicroRNAs. Endocrinology and metabolism clinics of North America. 2018;47(3):471-83.
  135. Mader S, Pantel K. Liquid Biopsy: Current Status and Future Perspectives. Oncology research and treatment. 2017;40(7-8):404-8.
  136. Boons G, Vandamme T, Peeters M, Beyens M, Driessen A, Janssens K, et al. Cell-Free DNA From Metastatic Pancreatic Neuroendocrine Tumor Patients Contains Tumor-Specific Mutations and Copy Number Variations. Frontiers in oncology. 2018;8:467.
  137. Malczewska A, Kidd M, Matar S, Kos-Kudla B, Modlin IM. A Comprehensive Assessment of the Role of miRNAs as Biomarkers in Gastroenteropancreatic Neuroendocrine Tumors. Neuroendocrinology. 2018;107(1):73-90.
  138. Li SC, Khan M, Caplin M, Meyer T, Öberg K, Giandomenico V. Somatostatin Analogs Treated Small Intestinal Neuroendocrine Tumor Patients Circulating MicroRNAs. PloS one. 2015;10(5):e0125553.
  139. Kövesdi A, Kurucz PA, Nyírő G, Darvasi O, Patócs A, Butz H. Circulating miRNA Increases the Diagnostic Accuracy of Chromogranin A in Metastatic Pancreatic Neuroendocrine Tumors. Cancers. 2020;12(9).
  140. Malczewska A, Kos-Kudła B, Kidd M, Drozdov I, Bodei L, Matar S, et al. The clinical applications of a multigene liquid biopsy (NETest) in neuroendocrine tumors. Advances in medical sciences. 2020;65(1):18-29.
  141. Modlin IM, Drozdov I, Kidd M. The identification of gut neuroendocrine tumor disease by multiple synchronous transcript analysis in blood. PloS one. 2013;8(5):e63364.
  142. Modlin IM, Kidd M, Falconi M, Filosso PL, Frilling A, Malczewska A, et al. A multigenomic liquid biopsy biomarker for neuroendocrine tumor disease outperforms CgA and has surgical and clinical utility. Annals of oncology : official journal of the European Society for Medical Oncology. 2021;32(11):1425-33.
  143. Modlin IM, Kidd M, Malczewska A, Drozdov I, Bodei L, Matar S, et al. The NETest: The Clinical Utility of Multigene Blood Analysis in the Diagnosis and Management of Neuroendocrine Tumors. Endocrinology and metabolism clinics of North America. 2018;47(3):485-504.
  144. Modlin IM, Frilling A, Salem RR, Alaimo D, Drymousis P, Wasan HS, et al. Blood measurement of neuroendocrine gene transcripts defines the effectiveness of operative resection and ablation strategies. Surgery. 2016;159(1):336-47.
  145. Ćwikła JB, Bodei L, Kolasinska-Ćwikła A, Sankowski A, Modlin IM, Kidd M. Circulating Transcript Analysis (NETest) in GEP-NETs Treated With Somatostatin Analogs Defines Therapy. The Journal of clinical endocrinology and metabolism. 2015;100(11):E1437-45.
  146. Bodei L, Kidd M, Modlin IM, Severi S, Drozdov I, Nicolini S, et al. Measurement of circulating transcripts and gene cluster analysis predicts and defines therapeutic efficacy of peptide receptor radionuclide therapy (PRRT) in neuroendocrine tumors. European journal of nuclear medicine and molecular imaging. 2016;43(5):839-51.
  147. van Treijen MJC, Korse CM, van Leeuwaarde RS, Saveur LJ, Vriens MR, Verbeek WHM, et al. Blood Transcript Profiling for the Detection of Neuroendocrine Tumors: Results of a Large Independent Validation Study. Frontiers in endocrinology. 2018;9:740.
  148. Lee L, Ito T, Jensen RT. Imaging of pancreatic neuroendocrine tumors: recent advances, current status, and controversies. Expert review of anticancer therapy. 2018;18(9):837-60.
  149. Akirov A, Larouche V, Alshehri S, Asa SL, Ezzat S. Treatment Options for Pancreatic Neuroendocrine Tumors. Cancers. 2019;11(6).
  150. Belotto M, Crouzillard B, Araujo KO, Peixoto RD. PANCREATIC NEUROENDOCRINE TUMORS: SURGICAL RESECTION. Arquivos brasileiros de cirurgia digestiva : ABCD = Brazilian archives of digestive surgery. 2019;32(1):e1428.
  151. Zerbi A, Capitanio V, Boninsegna L, Pasquali C, Rindi G, Delle Fave G, et al. Surgical treatment of pancreatic endocrine tumours in Italy: results of a prospective multicentre study of 262 cases. Langenbeck's archives of surgery. 2011;396(3):313-21.
  152. Hill JS, McPhee JT, McDade TP, Zhou Z, Sullivan ME, Whalen GF, et al. Pancreatic neuroendocrine tumors: the impact of surgical resection on survival. Cancer. 2009;115(4):741-51.
  153. Gomez-Rivera F, Stewart AE, Arnoletti JP, Vickers S, Bland KI, Heslin MJ. Surgical treatment of pancreatic endocrine neoplasms. American journal of surgery. 2007;193(4):460-5.
  154. Martin RC, Kooby DA, Weber SM, Merchant NB, Parikh AA, Cho CS, et al. Analysis of 6,747 pancreatic neuroendocrine tumors for a proposed staging system. Journal of gastrointestinal surgery : official journal of the Society for Surgery of the Alimentary Tract. 2011;15(1):175-83.
  155. Scarpa A, Mantovani W, Capelli P, Beghelli S, Boninsegna L, Bettini R, et al. Pancreatic endocrine tumors: improved TNM staging and histopathological grading permit a clinically efficient prognostic stratification of patients. Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc. 2010;23(6):824-33.
  156. Toste PA, Kadera BE, Tatishchev SF, Dawson DW, Clerkin BM, Muthusamy R, et al. Nonfunctional pancreatic neuroendocrine tumors <2 cm on preoperative imaging are associated with a low incidence of nodal metastasis and an excellent overall survival. Journal of gastrointestinal surgery : official journal of the Society for Surgery of the Alimentary Tract. 2013;17(12):2105-13.
  157. Partelli S, Gaujoux S, Boninsegna L, Cherif R, Crippa S, Couvelard A, et al. Pattern and clinical predictors of lymph node involvement in nonfunctioning pancreatic neuroendocrine tumors (NF-PanNETs). JAMA surgery. 2013;148(10):932-9.
  158. Titan AL, Norton JA, Fisher AT, Foster DS, Harris EJ, Worhunsky DJ, et al. Evaluation of Outcomes Following Surgery for Locally Advanced Pancreatic Neuroendocrine Tumors. JAMA network open. 2020;3(11):e2024318.
  159. Bettini R, Partelli S, Boninsegna L, Capelli P, Crippa S, Pederzoli P, et al. Tumor size correlates with malignancy in nonfunctioning pancreatic endocrine tumor. Surgery. 2011;150(1):75-82.
  160. Gaujoux S, Partelli S, Maire F, D'Onofrio M, Larroque B, Tamburrino D, et al. Observational study of natural history of small sporadic nonfunctioning pancreatic neuroendocrine tumors. The Journal of clinical endocrinology and metabolism. 2013;98(12):4784-9.
  161. Triponez F, Sadowski SM, Pattou F, Cardot-Bauters C, Mirallié E, Le Bras M, et al. Long-term Follow-up of MEN1 Patients Who Do Not Have Initial Surgery for Small ≤2 cm Nonfunctioning Pancreatic Neuroendocrine Tumors, an AFCE and GTE Study: Association Francophone de Chirurgie Endocrinienne & Groupe d'Etude des Tumeurs Endocrines. Annals of surgery. 2018;268(1):158-64.
  162. Thakker RV, Newey PJ, Walls GV, Bilezikian J, Dralle H, Ebeling PR, et al. Clinical practice guidelines for multiple endocrine neoplasia type 1 (MEN1). The Journal of clinical endocrinology and metabolism. 2012;97(9):2990-3011.
  163. Krug S, Gress TM, Michl P, Rinke A. The Role of Cytotoxic Chemotherapy in Advanced Pancreatic Neuroendocrine Tumors. Digestion. 2017;96(2):67-75.
  164. Akerström G, Hellman P. Surgery on neuroendocrine tumours. Best practice & research Clinical endocrinology & metabolism. 2007;21(1):87-109.
  165. Jensen RT, Norton JA. Treatment of Pancreatic Neuroendocrine Tumors in Multiple Endocrine Neoplasia Type 1: Some Clarity But Continued Controversy. Pancreas. 2017;46(5):589-94.
  166. Jensen RT, Niederle B, Mitry E, Ramage JK, Steinmuller T, Lewington V, et al. Gastrinoma (duodenal and pancreatic). Neuroendocrinology. 2006;84(3):173-82.
  167. Norton JA, Fraker DL, Alexander HR, Venzon DJ, Doppman JL, Serrano J, et al. Surgery to cure the Zollinger-Ellison syndrome. The New England journal of medicine. 1999;341(9):635-44.
  168. Al-Kurd A, Chapchay K, Grozinsky-Glasberg S, Mazeh H. Laparoscopic resection of pancreatic neuroendocrine tumors. World journal of gastroenterology. 2014;20(17):4908-16.
  169. Drymousis P, Raptis DA, Spalding D, Fernandez-Cruz L, Menon D, Breitenstein S, et al. Laparoscopic versus open pancreas resection for pancreatic neuroendocrine tumours: a systematic review and meta-analysis. HPB : the official journal of the International Hepato Pancreato Biliary Association. 2014;16(5):397-406.
  170. Venkat R, Edil BH, Schulick RD, Lidor AO, Makary MA, Wolfgang CL. Laparoscopic distal pancreatectomy is associated with significantly less overall morbidity compared to the open technique: a systematic review and meta-analysis. Annals of surgery. 2012;255(6):1048-59.
  171. Kendrick ML, van Hilst J, Boggi U, de Rooij T, Walsh RM, Zeh HJ, et al. Minimally invasive pancreatoduodenectomy. HPB : the official journal of the International Hepato Pancreato Biliary Association. 2017;19(3):215-24.
  172. Modlin IM, Lye KD, Kidd M. A 5-decade analysis of 13,715 carcinoid tumors. Cancer. 2003;97(4):934-59.
  173. Chamberlain RS, Canes D, Brown KT, Saltz L, Jarnagin W, Fong Y, et al. Hepatic neuroendocrine metastases: does intervention alter outcomes? Journal of the American College of Surgeons. 2000;190(4):432-45.
  174. Hodul PJ, Strosberg JR, Kvols LK. Aggressive surgical resection in the management of pancreatic neuroendocrine tumors: when is it indicated? Cancer control : journal of the Moffitt Cancer Center. 2008;15(4):314-21.
  175. Zhou B, Zhan C, Ding Y, Yan S, Zheng S. Role of palliative resection of the primary pancreatic neuroendocrine tumor in patients with unresectable metastatic liver disease: a systematic review and meta-analysis. OncoTargets and therapy. 2018;11:975-82.
  176. Mazzaferro V, Pulvirenti A, Coppa J. Neuroendocrine tumors metastatic to the liver: how to select patients for liver transplantation? Journal of hepatology. 2007;47(4):460-6.
  177. Shah MH, Goldner WS, Halfdanarson TR, Bergsland E, Berlin JD, Halperin D, et al. NCCN Guidelines Insights: Neuroendocrine and Adrenal Tumors, Version 2.2018. Journal of the National Comprehensive Cancer Network : JNCCN. 2018;16(6):693-702.
  178. Solorzano CC, Lee JE, Pisters PW, Vauthey JN, Ayers GD, Jean ME, et al. Nonfunctioning islet cell carcinoma of the pancreas: survival results in a contemporary series of 163 patients. Surgery. 2001;130(6):1078-85.
  179. Elias D, Lefevre JH, Duvillard P, Goéré D, Dromain C, Dumont F, et al. Hepatic metastases from neuroendocrine tumors with a "thin slice" pathological examination: they are many more than you think. Annals of surgery. 2010;251(2):307-10.
  180. Sarmiento JM, Heywood G, Rubin J, Ilstrup DM, Nagorney DM, Que FG. Surgical treatment of neuroendocrine metastases to the liver: a plea for resection to increase survival. Journal of the American College of Surgeons. 2003;197(1):29-37.
  181. Norton JA, Sugarbaker PH, Doppman JL, Wesley RA, Maton PN, Gardner JD, et al. Aggressive resection of metastatic disease in selected patients with malignant gastrinoma. Annals of surgery. 1986;203(4):352-9.
  182. Sarmiento JM, Que FG. Hepatic surgery for metastases from neuroendocrine tumors. Surgical oncology clinics of North America. 2003;12(1):231-42.
  183. House MG, Cameron JL, Lillemoe KD, Schulick RD, Choti MA, Hansel DE, et al. Differences in survival for patients with resectable versus unresectable metastases from pancreatic islet cell cancer. Journal of gastrointestinal surgery : official journal of the Society for Surgery of the Alimentary Tract. 2006;10(1):138-45.
  184. Gupta S, Johnson MM, Murthy R, Ahrar K, Wallace MJ, Madoff DC, et al. Hepatic arterial embolization and chemoembolization for the treatment of patients with metastatic neuroendocrine tumors: variables affecting response rates and survival. Cancer. 2005;104(8):1590-602.
  185. Mazzaferro V, Sposito C, Coppa J, Miceli R, Bhoori S, Bongini M, et al. The Long-Term Benefit of Liver Transplantation for Hepatic Metastases From Neuroendocrine Tumors. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2016;16(10):2892-902.
  186. Herrera-Martínez AD, Hofland J, Hofland LJ, Brabander T, Eskens F, Gálvez Moreno MA, et al. Targeted Systemic Treatment of Neuroendocrine Tumors: Current Options and Future Perspectives. Drugs. 2019;79(1):21-42.
  187. Gomes-Porras M, Cárdenas-Salas J, Álvarez-Escolá C. Somatostatin Analogs in Clinical Practice: a Review. International journal of molecular sciences. 2020;21(5).
  188. Hauser H, Gerson DS, Reidy-Lagunes D, Raj N. Systemic Therapies for Metastatic Pancreatic Neuroendocrine Tumors. Current treatment options in oncology. 2019;20(12):87.
  189. Caplin ME, Pavel M, Ćwikła JB, Phan AT, Raderer M, Sedláčková E, et al. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. The New England journal of medicine. 2014;371(3):224-33.
  190. Caplin ME, Pavel M, Ćwikła JB, Phan AT, Raderer M, Sedláčková E, et al. Anti-tumour effects of lanreotide for pancreatic and intestinal neuroendocrine tumours: the CLARINET open-label extension study. Endocrine-related cancer. 2016;23(3):191-9.
  191. Pavel M, Öberg K, Falconi M, Krenning EP, Sundin A, Perren A, et al. Gastroenteropancreatic neuroendocrine neoplasms: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Annals of oncology : official journal of the European Society for Medical Oncology. 2020;31(7):844-60.
  192. Vinik AI, Raymond E. Pancreatic neuroendocrine tumors: approach to treatment with focus on sunitinib. Therapeutic advances in gastroenterology. 2013;6(5):396-411.
  193. Kulke MH, Lenz HJ, Meropol NJ, Posey J, Ryan DP, Picus J, et al. Activity of sunitinib in patients with advanced neuroendocrine tumors. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2008;26(20):3403-10.
  194. Raymond E, Dahan L, Raoul JL, Bang YJ, Borbath I, Lombard-Bohas C, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. The New England journal of medicine. 2011;364(6):501-13.
  195. Faivre S, Niccoli P, Castellano D, Valle JW, Hammel P, Raoul JL, et al. Sunitinib in pancreatic neuroendocrine tumors: updated progression-free survival and final overall survival from a phase III randomized study. Annals of oncology : official journal of the European Society for Medical Oncology. 2017;28(2):339-43.
  196. Vinik E, Carlton CA, Silva MP, Vinik AI. Development of the Norfolk quality of life tool for assessing patients with neuroendocrine tumors. Pancreas. 2009;38(3):e87-95.
  197. Yao JC, Lombard-Bohas C, Baudin E, Kvols LK, Rougier P, Ruszniewski P, et al. Daily oral everolimus activity in patients with metastatic pancreatic neuroendocrine tumors after failure of cytotoxic chemotherapy: a phase II trial. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2010;28(1):69-76.
  198. Yao JC, Shah MH, Ito T, Bohas CL, Wolin EM, Van Cutsem E, et al. Everolimus for advanced pancreatic neuroendocrine tumors. The New England journal of medicine. 2011;364(6):514-23.
  199. Lombard-Bohas C, Yao JC, Hobday T, Van Cutsem E, Wolin EM, Panneerselvam A, et al. Impact of prior chemotherapy use on the efficacy of everolimus in patients with advanced pancreatic neuroendocrine tumors: a subgroup analysis of the phase III RADIANT-3 trial. Pancreas. 2015;44(2):181-9.
  200. Chan JA, Blaszkowsky L, Stuart K, Zhu AX, Allen J, Wadlow R, et al. A prospective, phase 1/2 study of everolimus and temozolomide in patients with advanced pancreatic neuroendocrine tumor. Cancer. 2013;119(17):3212-8.
  201. Kulke MH, Ruszniewski P, Van Cutsem E, Lombard-Bohas C, Valle JW, De Herder WW, et al. A randomized, open-label, phase 2 study of everolimus in combination with pasireotide LAR or everolimus alone in advanced, well-differentiated, progressive pancreatic neuroendocrine tumors: COOPERATE-2 trial. Annals of oncology : official journal of the European Society for Medical Oncology. 2017;28(6):1309-15.
  202. Halfdanarson TR, Strosberg JR, Tang L, Bellizzi AM, Bergsland EK, O'Dorisio TM, et al. The North American Neuroendocrine Tumor Society Consensus Guidelines for Surveillance and Medical Management of Pancreatic Neuroendocrine Tumors. Pancreas. 2020;49(7):863-81.
  203. Yao J, Phan AT. Optimising Therapeutic Options for Patients with Advanced Pancreatic Neuroendocrine Tumours. European Oncology & Haematology. 2012;8(4):217-23.
  204. Mateo J, Heymach JV, Zurita AJ. Circulating biomarkers of response to sunitinib in gastroenteropancreatic neuroendocrine tumors: current data and clinical outlook. Molecular diagnosis & therapy. 2012;16(3):151-61.
  205. Bello C, Deprimo SE, Friece C, Smeraglia J, Sherman L, Tye L, et al. Analysis of circulating biomarkers of sunitinib malate in patients with unresectable neuroendocrine tumors (NET): VEGF, IL-8, and soluble VEGF receptors 2 and 3. Journal of Clinical Oncology. 2006;24(18_suppl):4045-.
  206. Yao JC, Pavel M, Phan AT, Kulke MH, Hoosen S, St Peter J, et al. Chromogranin A and neuron-specific enolase as prognostic markers in patients with advanced pNET treated with everolimus. The Journal of clinical endocrinology and metabolism. 2011;96(12):3741-9.
  207. Palmieri LJ, Dermine S, Barré A, Dhooge M, Brezault C, Cottereau AS, et al. Medical Treatment of Advanced Pancreatic Neuroendocrine Neoplasms. Journal of clinical medicine. 2020;9(6).
  208. Pavel M, O'Toole D, Costa F, Capdevila J, Gross D, Kianmanesh R, et al. ENETS Consensus Guidelines Update for the Management of Distant Metastatic Disease of Intestinal, Pancreatic, Bronchial Neuroendocrine Neoplasms (NEN) and NEN of Unknown Primary Site. Neuroendocrinology. 2016;103(2):172-85.
  209. Okusaka T, Ueno H, Morizane C, Kondo S, Sakamoto Y, Takahashi H, et al. Cytotoxic chemotherapy for pancreatic neuroendocrine tumors. Journal of hepato-biliary-pancreatic sciences. 2015;22(8):628-33.
  210. Moertel CG, Lefkopoulo M, Lipsitz S, Hahn RG, Klaassen D. Streptozocin-doxorubicin, streptozocin-fluorouracil or chlorozotocin in the treatment of advanced islet-cell carcinoma. The New England journal of medicine. 1992;326(8):519-23.
  211. Kouvaraki MA, Ajani JA, Hoff P, Wolff R, Evans DB, Lozano R, et al. Fluorouracil, doxorubicin, and streptozocin in the treatment of patients with locally advanced and metastatic pancreatic endocrine carcinomas. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2004;22(23):4762-71.
  212. Ramanathan RK, Cnaan A, Hahn RG, Carbone PP, Haller DG. Phase II trial of dacarbazine (DTIC) in advanced pancreatic islet cell carcinoma. Study of the Eastern Cooperative Oncology Group-E6282. Annals of oncology : official journal of the European Society for Medical Oncology. 2001;12(8):1139-43.
  213. de Mestier L, Walter T, Brixi H, Evrard C, Legoux JL, de Boissieu P, et al. Comparison of Temozolomide-Capecitabine to 5-Fluorouracile-Dacarbazine in 247 Patients with Advanced Digestive Neuroendocrine Tumors Using Propensity Score Analyses. Neuroendocrinology. 2019;108(4):343-53.
  214. Strosberg JR, Fine RL, Choi J, Nasir A, Coppola D, Chen DT, et al. First-line chemotherapy with capecitabine and temozolomide in patients with metastatic pancreatic endocrine carcinomas. Cancer. 2011;117(2):268-75.
  215. Chan JA, Stuart K, Earle CC, Clark JW, Bhargava P, Miksad R, et al. Prospective study of bevacizumab plus temozolomide in patients with advanced neuroendocrine tumors. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2012;30(24):2963-8.
  216. Koumarianou A, Antoniou S, Kanakis G, Economopoulos N, Rontogianni D, Ntavatzikos A, et al. Combination treatment with metronomic temozolomide, bevacizumab and long-acting octreotide for malignant neuroendocrine tumours. Endocrine-related cancer. 2012;19(1):L1-4.
  217. Kulke MH, Stuart K, Enzinger PC, Ryan DP, Clark JW, Muzikansky A, et al. Phase II study of temozolomide and thalidomide in patients with metastatic neuroendocrine tumors. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2006;24(3):401-6.
  218. Kunz PL, Catalano PJ, Nimeiri H, Fisher GA, Longacre TA, Suarez CJ, et al. A randomized study of temozolomide or temozolomide and capecitabine in patients with advanced pancreatic neuroendocrine tumors: A trial of the ECOG-ACRIN Cancer Research Group (E2211). Journal of Clinical Oncology. 2018;36(15_suppl):4004-.
  219. de Mestier L, Walter T, Evrard C, de Boissieu P, Hentic O, Cros J, et al. Temozolomide Alone or Combined with Capecitabine for the Treatment of Advanced Pancreatic Neuroendocrine Tumor. Neuroendocrinology. 2020;110(1-2):83-91.
  220. Moertel CG, Kvols LK, O'Connell MJ, Rubin J. Treatment of neuroendocrine carcinomas with combined etoposide and cisplatin. Evidence of major therapeutic activity in the anaplastic variants of these neoplasms. Cancer. 1991;68(2):227-32.
  221. Mitry E, Baudin E, Ducreux M, Sabourin JC, Rufié P, Aparicio T, et al. Treatment of poorly differentiated neuroendocrine tumours with etoposide and cisplatin. British journal of cancer. 1999;81(8):1351-5.
  222. Thomas KEH, Voros BA, Boudreaux JP, Thiagarajan R, Woltering EA, Ramirez RA. Current Treatment Options in Gastroenteropancreatic Neuroendocrine Carcinoma. The oncologist. 2019;24(8):1076-88.
  223. Ilett EE, Langer SW, Olsen IH, Federspiel B, Kjær A, Knigge U. Neuroendocrine Carcinomas of the Gastroenteropancreatic System: A Comprehensive Review. Diagnostics (Basel, Switzerland). 2015;5(2):119-76.
  224. Sorbye H, Welin S, Langer SW, Vestermark LW, Holt N, Osterlund P, et al. Predictive and prognostic factors for treatment and survival in 305 patients with advanced gastrointestinal neuroendocrine carcinoma (WHO G3): the NORDIC NEC study. Annals of oncology : official journal of the European Society for Medical Oncology. 2013;24(1):152-60.
  225. Hentic O, Hammel P, Couvelard A, Rebours V, Zappa M, Palazzo M, et al. FOLFIRI regimen: an effective second-line chemotherapy after failure of etoposide-platinum combination in patients with neuroendocrine carcinomas grade 3. Endocrine-related cancer. 2012;19(6):751-7.
  226. Bajetta E, Catena L, Procopio G, De Dosso S, Bichisao E, Ferrari L, et al. Are capecitabine and oxaliplatin (XELOX) suitable treatments for progressing low-grade and high-grade neuroendocrine tumours? Cancer chemotherapy and pharmacology. 2007;59(5):637-42.
  227. Spada F, Antonuzzo L, Marconcini R, Radice D, Antonuzzo A, Ricci S, et al. Oxaliplatin-Based Chemotherapy in Advanced Neuroendocrine Tumors: Clinical Outcomes and Preliminary Correlation with Biological Factors. Neuroendocrinology. 2016;103(6):806-14.
  228. Zhu J, Strosberg JR, Dropkin E, Strickler JH. Treatment of High-Grade Metastatic Pancreatic Neuroendocrine Carcinoma with FOLFIRINOX. Journal of gastrointestinal cancer. 2015;46(2):166-9.
  229. Camus B, Cottereau AS, Palmieri LJ, Dermine S, Tenenbaum F, Brezault C, et al. Indications of Peptide Receptor Radionuclide Therapy (PRRT) in Gastroenteropancreatic and Pulmonary Neuroendocrine Tumors: An Updated Review. Journal of clinical medicine. 2021;10(6).
  230. Kwekkeboom DJ, Teunissen JJ, Bakker WH, Kooij PP, de Herder WW, Feelders RA, et al. Radiolabeled somatostatin analog [177Lu-DOTA0,Tyr3]octreotate in patients with endocrine gastroenteropancreatic tumors. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2005;23(12):2754-62.
  231. Vinik E, Silva MP, Vinik AI. Measuring the relationship of quality of life and health status, including tumor burden, symptoms, and biochemical measures in patients with neuroendocrine tumors. Endocrinology and metabolism clinics of North America. 2011;40(1):97-109, viii.
  232. Ezziddin S, Khalaf F, Vanezi M, Haslerud T, Mayer K, Al Zreiqat A, et al. Outcome of peptide receptor radionuclide therapy with 177Lu-octreotate in advanced grade 1/2 pancreatic neuroendocrine tumours. European journal of nuclear medicine and molecular imaging. 2014;41(5):925-33.
  233. Strosberg J, El-Haddad G, Wolin E, Hendifar A, Yao J, Chasen B, et al. Phase 3 Trial of (177)Lu-Dotatate for Midgut Neuroendocrine Tumors. The New England journal of medicine. 2017;376(2):125-35.
  234. Food and Drug Administration. FDA approves lutetium Lu 177 dotatate for treatment of GEP-NETS. 2018.
  235. Bodei L, Mueller-Brand J, Baum RP, Pavel ME, Hörsch D, O'Dorisio MS, et al. The joint IAEA, EANM, and SNMMI practical guidance on peptide receptor radionuclide therapy (PRRNT) in neuroendocrine tumours. European journal of nuclear medicine and molecular imaging. 2013;40(5):800-16.
  236. Brabander T, van der Zwan WA, Teunissen JJM, Kam BLR, Feelders RA, de Herder WW, et al. Long-Term Efficacy, Survival, and Safety of [(177)Lu-DOTA(0),Tyr(3)]octreotate in Patients with Gastroenteropancreatic and Bronchial Neuroendocrine Tumors. Clinical cancer research : an official journal of the American Association for Cancer Research. 2017;23(16):4617-24.
  237. Satapathy S, Mittal BR. 177Lu-DOTATATE peptide receptor radionuclide therapy versus Everolimus in advanced pancreatic neuroendocrine tumors: a systematic review and meta-analysis. Nuclear medicine communications. 2019;40(12):1195-203.
  238. Pusceddu S, Prinzi N, Tafuto S, Ibrahim T, Filice A, Brizzi MP, et al. Association of Upfront Peptide Receptor Radionuclide Therapy With Progression-Free Survival Among Patients With Enteropancreatic Neuroendocrine Tumors. JAMA network open. 2022;5(2):e220290.
  239. Howe JR, Merchant NB, Conrad C, Keutgen XM, Hallet J, Drebin JA, et al. The North American Neuroendocrine Tumor Society Consensus Paper on the Surgical Management of Pancreatic Neuroendocrine Tumors. Pancreas. 2020;49(1):1-33.
  240. Bloomston M, Al-Saif O, Klemanski D, Pinzone JJ, Martin EW, Palmer B, et al. Hepatic artery chemoembolization in 122 patients with metastatic carcinoid tumor: lessons learned. Journal of gastrointestinal surgery : official journal of the Society for Surgery of the Alimentary Tract. 2007;11(3):264-71.
  241. Kennedy AS, Dezarn WA, McNeillie P, Coldwell D, Nutting C, Carter D, et al. Radioembolization for unresectable neuroendocrine hepatic metastases using resin 90Y-microspheres: early results in 148 patients. American journal of clinical oncology. 2008;31(3):271-9.
  242. Rhee TK, Lewandowski RJ, Liu DM, Mulcahy MF, Takahashi G, Hansen PD, et al. 90Y Radioembolization for metastatic neuroendocrine liver tumors: preliminary results from a multi-institutional experience. Annals of surgery. 2008;247(6):1029-35.
  243. Grozinsky-Glasberg S, Kaltsas G, Kaltsatou M, Lev-Cohain N, Klimov A, Vergadis V, et al. Hepatic intra-arterial therapies in metastatic neuroendocrine tumors: lessons from clinical practice. Endocrine. 2018;60(3):499-509.
  244. Patnaik A, Kang SP, Rasco D, Papadopoulos KP, Elassaiss-Schaap J, Beeram M, et al. Phase I Study of Pembrolizumab (MK-3475; Anti-PD-1 Monoclonal Antibody) in Patients with Advanced Solid Tumors. Clinical cancer research : an official journal of the American Association for Cancer Research. 2015;21(19):4286-93.
  245. McGranahan N, Furness AJ, Rosenthal R, Ramskov S, Lyngaa R, Saini SK, et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science (New York, NY). 2016;351(6280):1463-9.
  246. Mehnert JM, Bergsland E, O'Neil BH, Santoro A, Schellens JHM, Cohen RB, et al. Pembrolizumab for the treatment of programmed death-ligand 1-positive advanced carcinoid or pancreatic neuroendocrine tumors: Results from the KEYNOTE-028 study. Cancer. 2020;126(13):3021-30.
  247. Strosberg J, Mizuno N, Doi T, Grande E, Delord JP, Shapira-Frommer R, et al. Efficacy and Safety of Pembrolizumab in Previously Treated Advanced Neuroendocrine Tumors: Results From the Phase II KEYNOTE-158 Study. Clinical cancer research : an official journal of the American Association for Cancer Research. 2020;26(9):2124-30.
  248. Naing A, Papadopoulos KP, Autio KA, Ott PA, Patel MR, Wong DJ, et al. Safety, Antitumor Activity, and Immune Activation of Pegylated Recombinant Human Interleukin-10 (AM0010) in Patients With Advanced Solid Tumors. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2016;34(29):3562-9.
  249. Yao JC, Guthrie KA, Moran C, Strosberg JR, Kulke MH, Chan JA, et al. Phase III Prospective Randomized Comparison Trial of Depot Octreotide Plus Interferon Alfa-2b Versus Depot Octreotide Plus Bevacizumab in Patients With Advanced Carcinoid Tumors: SWOG S0518. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2017;35(15):1695-703.
  250. Ducreux M, Dahan L, Smith D, O'Toole D, Lepère C, Dromain C, et al. Bevacizumab combined with 5-FU/streptozocin in patients with progressive metastatic well-differentiated pancreatic endocrine tumours (BETTER trial)--a phase II non-randomised trial. European journal of cancer (Oxford, England : 1990). 2014;50(18):3098-106.
  251. Xu J, Shen L, Zhou Z, Li J, Bai C, Chi Y, et al. Surufatinib in advanced extrapancreatic neuroendocrine tumours (SANET-ep): a randomised, double-blind, placebo-controlled, phase 3 study. The Lancet Oncology. 2020;21(11):1500-12.
  252. Xu J, Shen L, Bai C, Wang W, Li J, Yu X, et al. Surufatinib in advanced pancreatic neuroendocrine tumours (SANET-p): a randomised, double-blind, placebo-controlled, phase 3 study. The Lancet Oncology. 2020;21(11):1489-99.
  253. Aaronson NK, Ahmedzai S, Bergman B, Bullinger M, Cull A, Duez NJ, et al. The European Organization for Research and Treatment of Cancer QLQ-C30: a quality-of-life instrument for use in international clinical trials in oncology. Journal of the National Cancer Institute. 1993;85(5):365-76.
  254. Yadegarfar G, Friend L, Jones L, Plum LM, Ardill J, Taal B, et al. Validation of the EORTC QLQ-GINET21 questionnaire for assessing quality of life of patients with gastrointestinal neuroendocrine tumours. British journal of cancer. 2013;108(2):301-10.
  255. Jiménez-Fonseca P, Carmona-Bayonas A, Martín-Pérez E, Crespo G, Serrano R, Llanos M, et al. Health-related quality of life in well-differentiated metastatic gastroenteropancreatic neuroendocrine tumors. Cancer metastasis reviews. 2015;34(3):381-400.
  256. Meng Y, McCarthy G, Berthon A, Dinet J. Patient-reported health state utilities in metastatic gastroenteropancreatic neuroendocrine tumours - an analysis based on the CLARINET study. Health and quality of life outcomes. 2017;15(1):131.
  257. Marinova M, Mücke M, Mahlberg L, Essler M, Cuhls H, Radbruch L, et al. Improving quality of life in patients with pancreatic neuroendocrine tumor following peptide receptor radionuclide therapy assessed by EORTC QLQ-C30. European journal of nuclear medicine and molecular imaging. 2018;45(1):38-46.
  258. Martini C, Gamper EM, Wintner L, Nilica B, Sperner-Unterweger B, Holzner B, et al. Systematic review reveals lack of quality in reporting health-related quality of life in patients with gastroenteropancreatic neuroendocrine tumours. Health and quality of life outcomes. 2016;14(1):127.