Archives

Adrenocortical Carcinoma

CLINICAL RECOGNITION

 

Adrenocortical cancer (ACC) is a rare disease with an annual incidence of 0.7-2 cases per million per year and two distinct age distribution peaks, the first occurring in early adulthood and the second between 40-50 years with women being more often affected (55-60%). Although the great majority of ACCs are of sporadic origin, they can also develop as part of familial diseases the most common being the Beckwith-Wiedeman syndrome, the Li-Fraumeni syndrome, the Lynch syndrome, the multiple endocrine neoplasia (MEN) 1, and familial adenomatous polyposis (FAP) (Table 1). In recent years several multi-center studies have shed light on the pathogenesis of ACC
but ‘multi-omic’ studies reveal that only a minority of ACC cases harbour pathogenic driver mutations.

 

Table 1. Clinical and Genetic Features of Familial Syndromes Associated with ACC

Genetic disease

Gene and chromosomal involvement

Organ involvement
Beckwith-Wiedemann syndrome

CDKN1C mutation

KCNQ10T1, H19 (epigenetic defects)

11p15 locus alterations

IGF-2 overexpression

Macrosomia, macroglossia, hemihypertrophy (70%), omphalocele, Wilm’s tumor, ACC (15-20% adrenocortical tumors)
Li-Fraumeni syndrome P53(17p13) Soft tissue sarcoma, breast cancer, brain tumors, leukemia, ACC
Multiple Endocrine Neoplasia syndrome 1

Menin (11q13)

Parathyroid, pituitary, pancreatic, bronchial tumors

Adrenal cortex tumors (30%, rarely ACC)

Familial Adenomatous polyposis

APC (5q12-22)

Multiple adenomatous polyps and cancer colon and rectum

Periampullary cancer, thyroid tumors, hepatoblastoma, rarely ACC

SBLA syndrome

 

Sarcoma, breast and lung cancer, ACC

 

The clinical features of sporadic ACCs are due to hormone hypersecretion and/or tumor mass and spread to surrounding or distant tissues. An increasing number of cases (≈ 10-15%) are increasingly been diagnosed within the group of incidentally discovered adrenal masses (incidentalomas). However, the likelihood of an adrenal incidentaloma being an ACC is rather low. Approximately 50-60% of ACCs exhibit evidence of hormonal hypersecretion, usually that of combined glucocorticoid and androgen secretion (Table 2). Nearly 30-40% of patients with primary ACC present with a mass syndrome as abdominal or dorsal pain, a palpable mass, fever of unknown origin, signs of inferior vena cava (IVC) compression, and signs of left-sided portal hypertension. Rarely, complications as hemorrhage or tumor rupture may also develop. Lately the number of patients that are identified while being investigated for an adrenal incidentaloma is rapidly increasing.

 

Symptoms/Signs Hormonal testing (ENSAT 2005)
Hypercortisolism

Centripetal fat distribution

Skin thinning – striae

Muscle wasting – myopathy

Osteoporosis

Increased blood pressure (BP)

Diabetes Mellitus

Psychiatric disturbance

Gonadal dysfunction

Overnight dexamethasone

suppression test (1mg)

24-hour free cortisol

Basal ACTH (plasma)

Basal cortisol (serum)

[for diagnosis minimum 3 out of 4 tests)

Androgen hypersecretion

Hirsutism

Menstrual irregularity – infertility

Virilization (baldness, deepening of the voice, clitoris hypertrophy)

DHEA-S

Androstendione

Testosterone

17-OH-progesterone

Mineralocorticoid hypersecretion

Mineralocorticoid excess with increased BP, hypokalemia

Potassium (serum)

Aldosterone to renin ratio

 

Estrogen hypersecretion

Gynecomastia (men)

Menorrhagia (post-menopausal women)

17β-estradiol
Non-hypersecretory syndrome

 

PATHOPHYSIOLOGY

 

Although studies of hereditary neoplasia syndromes have revealed various chromosomal abnormalities related to ACC development the precise genetic alterations involved are still unknown. The Wnt/β-catenin constitutive activation and insulin growth factor 2 (IGF2 overexpression) are the most important implicated genetic pathways.  Germline TP53 mutations and dysregulation of the Gap 2/mitosis transition and the insulin-like growth factor 1 receptor (IGF1R) signalling have also been described. Steroidogenic factor 1 (SF1) plays an important role in adrenal development and is frequently overexpressed in ACC.

 

DIAGNOSIS AND DIFFERENTIAL DIAGNOSIS

 

A palpable mass causing abdominal pain in the presence of the inferior vena cava syndrome IVC syndrome is highly suggestive of an ACC. This is substantiated further by the presence of symptoms/signs of combined hormonal secretion (cortisol and androgens), virilizing or rarely feminizing signs confirmed with the use of specific endocrine testing (Table 2). As the majority of ACCs are relatively large (size > 8cm, weight >100g) at diagnosis, specific imaging features are used to distinguish them from other adrenal lesions. If adrenal imaging indicates an indeterminate mass other parameters should be considered including tumor size > 4 cm, combined cortisol/androgen hormone excess, rapidly developing symptoms and/ rapid tumor growth and/or young age (e.g. < 40 years) that might point to an ACC.

 

Other adrenal lesions that need to be considered in the differential diagnosis are myelolipomas, adrenal hemorrhage, lymphoma, adrenal cysts, metastases, and mainly adrenal adenomas, the majority of which have distinctive imaging features. There is no role for biopsy in a patient who is considered suitable for surgery of the adrenal mass.

 

Computerized Tomography (CT) scanning of the adrenals is the major tool showing a unilateral non-homogenous mass, >5cm in diameter, with irregular margins, necrosis, and occasionally calcifications. Due to the low-fat content X-Ray density is high (>20 Hounsfield Units, HU); in a recent series of 51 ACC none had a density of less than 13 HU. The presence of enlarged aorto-caval lymph nodes, local invasion, or metastatic spread are highly suggestive of ACC. For 3-6 cm size lesions, measuring X-Ray tumoral density before and after contrast administration and estimating washout percentage can be helpful; less than 50% after 15 minutes, is associated with >90% specificity. On Magnetic Resonance Imaging (MRI), ACC appears hypo or isointense to the liver on T1-weighted images, and using gadolinium enhancement and chemical shift techniques the diagnostic accuracy obtained is 85-100%. Recently Positron Emission Tomography (PET-scan) with 18F-fluoro-2deoxy-D-glucose (18FDG) has been proposed as possibly the best second-line test to assess indeterminate masses by unenhanced CT exhibiting 95-100% sensitivity and 91-94% specificity that increases further when fused with CT imaging. Furthermore, 18FDG-PET can also be used as a staging procedure identifying metastatic adrenal disease missed by conventional imaging studies including CT of the chest. With the proper implementation of imaging studies there is no need for adrenal biopsy.

 

HISTOPATHOLOGICAL DIAGNOSIS

 

The expression of SF1 is a valid marker to document the adrenal origin (distinction of primary adrenocortical tumors and non-adrenocortical tumors) with a sensitivity of 98% and a specificity of 100%. If this marker is not available, a combination of other markers can be used which should include inhibin-alpha, melan-A, and calretinin. ENSAT has shown that KI67 is the most powerful prognostic marker in both localized and advanced ACC and that higher Ki67 levels are consistently associated with worse prognosis. Weiss system, based on a combination of 9 histological criteria that can be applied on hematoxylin and eosin-stained slides, for the distinction of benign and malignant adrenocortical tumors is the best validated score to distinguish adenomas from ACC although with high inter-observer variability.

 

PROGNOSIS

 

As survival depends on stage at presentation several different classification histopathological systems have evolved with the reported 5-year survival using the ENSAT system being 82% for stage I, 61% for stage II, 50% for stage III, and 13% for stage IV (Table 3). Tumor size remains an excellent predictor of malignancy as tumors > 6cm have a 25% chance of being malignant compared to 2% of those with a size < 4cm. As there is no single distinctive histopathological feature indicative of malignancy the Weiss score has been used with a score >3 being suggestive of malignancy and recently ki67 labelling index >10%.

 

Table 3. Staging system for adrenocortical carcinomas proposed by the International Union against cancer (WHO 2004) and the European Network for the study of adrenal tumors (ENSAT).

Stage WHO 2004 ENSAT 2008
I T1,N0,M0 T1,N0,M0
II T2,N0,M0 T2,N0,M0
III T1-2,N1,M0

T3,N0,M0

T1-2,N1,M0
IV T1-4,N0-1,M1

T3,N1,M0

T4,N0-1,M0

T1-4,N0-1, M1
M0: No distant metastasis, M1: Presence of distant metastasis, N0: No positive lymph nodes, N1: Positive lymph node(s), T1: Tumor ≤ 5cm, T2: Tumor > 5 cm, T3: Tumor infiltration to surrounding tissue, T4: Tumor invasion into adjacent organs or venous tumor thrombus in vena cava or renal vein.

 

The median overall survival (OS) of all ACC patients is about 3-4 years. The prognosis is, however, heterogeneous. Complete surgical resection provides the only means of cure. In addition to radical surgery, disease stage, proliferative activity/tumor grade, and cortisol excess are independent prognostic parameters. Five-year survival rate is 60-80% for tumors confined to the adrenal space, 35-50% for locally advanced disease, and significantly lower in case of metastatic disease ranging from 0% to 28%. European Network for the Study of Adrenal Tumors (ENSAT) staging is considered slightly superior to the Union for International Cancer Control (UICC) staging. Additionally, the association between hypercortisolism and mortality was consistent. As Ki67 has been shown to be related with prognosis in both localized and advanced ACC threshold levels of 10% and 20% have been considered for discriminating low from high Ki67 labelling index; however, it is not clear whether any single significant threshold can be determined. Patients with stage I-III disease treated with surgical resection had significantly better median OS (63 vs. 8 months; p= 0.001). In stage IV disease, better median OS occurred in patients treated with surgery (19 vs. 6 months; p=0.001), and postsurgical radiation (29 vs 10 months; p=0.001) or chemotherapy (22 vs. 13 months; p= 0.004). Overall survival varied with increasing age, higher comorbidity index, grade, and stage of ACC at presentation. There was improved survival with surgical resection of the primary tumor, irrespective of disease stage; post-surgical chemotherapy or radiation was of benefit only in stage IV disease.

 

THERAPY

 

The management of patients with ACC requires a multidisciplinary approach with initial complete surgical resection in limited disease (stage I, II and occasionally III). Mitotane (1,1-dichloro-2(o-chlorophenyl)-2-(p-chlorophenyl) ethane [o,p’DDD]) is the only currently available adrenolytic medication achieving an overall response of approximately 30%.

Surgery

 

The aim of surgery is to achieve a complete margin-negative (R0) resection as patients with an R0 resection have a 5-year survival rate of 40-50% compared to the < 1year survival of those with incomplete resection. Patients with stage III tumors and positive lymph nodes can have a 10-year OS rate of up to 40% after complete resection. When a preoperative diagnosis or high level of suspicion of ACC exists, open surgical oncological resection is recommended as locoregional lymph removal might improve diagnostic accuracy and therapeutic outcome. However, the wide range of reported lymph node involvement in ACC (from 4 to 73%) implies that regional lymphadenectomy is neither formally performed by all surgeons nor accurately assessed or reported by all pathologists. Laparoscopic adrenalectomy should be considered for tumors with size up to 6 cm without any evidence of local invasion. Routine locoregional lymphadenectomy should be performed with adrenalectomy for highly suspected or proven ACC and it should include (as a minimum) the peri-adrenal and renal hilum nodes.

Preservation of the tumor capsule is essential whereas involvement of the IVC or renal vein with tumor thrombus is not a contraindication for surgery. However, even following an apparently complete surgical resection, 50-80% of patients develop locoregional or metastatic recurrence. Although such patients may be candidates for aggressive surgical resection, routine debulking is not recommended except for control of hormonal hypersecretion. Ablative therapies particularly targeting hepatic disease are used to decrease tumor load and the hypersecretory syndromes. Individualized treatment decisions are made in cases of tumors with extension into large vessels based on multidisciplinary surgical team. Such tumors should not be regarded ‘un-resectable’ until reviewed in an expert center.

Mitotane

 

Mitotane has traditionally been used for ACCs obtaining a partial or complete response in 33% of cases mainly by metabolic transformation within the tumor and through oxidative damage. Besides its cytotoxic adrenal action mitotane also inhibits steroidogenesis.

 

Adjuvant mitotane treatment is proposed in those patients without macroscopic residual tumor after surgery but who have a perceived high risk of recurrence (stage III, KI-67%>10%). However, for patients at low/moderate risk of recurrence (stage I-II, R0 resection, and Ki67 ≤ 10%) treatment with adjunct mitotane is still under investigation (results from ADIUVO trial are pending). When indicated mitotane should be initiated within six weeks and not later than 3 months. Adjuvant mitotane should be administrated for at least 2 years, but no longer than 5 years.

 

The tolerability of mitotane may be limited by its side effects mainly nausea, vomiting, neurological (ataxia, lethargy), hepatic and rarely hematological toxicity. Measurement of serum mitotane levels, targeting a range of 14-20 mg/l, seems to correlate with a therapeutic response while minimizing toxicity using variable dosing regimens. Mitotane causes hyperlipidemia and increased hepatic production of hormone binding globulins (cortisol, sex hormone, thyroid and vitamin D) increasing total hormone concentration while impairing free hormone bioavailability. The induction of hepatic P450-enzymes by mitotane induces the metabolism of steroid compounds requiring high dose glucocorticoid and mineralocorticoid replacement.

 

Hormonal excess causes significant morbidity in ACC patients. Although mitotane reduces steroidogenesis it has a slow onset of action necessitating the use of other adrenostatic medications (ketoconazole, metyrapone, aminoglutathemide, and etomidate). As adrenal insufficiency may occur close supervision is required to titrate adrenal hormonal replacement therapy.

Cytotoxic Chemotherapy

 

Although cisplatin containing regimens have shown some responses most studies lack power and comparisons between different regimens. The most encouraging results originate from the combinations of etoposide, doxorubicin and cisplatin with mitotane (EDP-M) achieving an overall response of 49% of 18 months duration (FIRMA-CT study). This regimen was equally effective as first line treatment or after failing of the combination of streptozotocin with mitotane and is the currently the preferred scheme. In patients who progress under mitotane monotherapy, EDP treatment is also recommended. The combination of gemcitabine with capecitabine is used for patients failing EDP- and for not responding patients targeted therapies with tyrosine kinase inhibitors (mainly sunitinib) could be used. Although initially promising treatment with IGF-1R antagonists did not prove to be efficacious suggesting that combination of therapies may be the way forward.

Radiation Therapy

 

Radiotherapy has a role in symptomatic metastatic disease particularly bone disease with positive responses in up to 50% - 90% of cancer patients.

Evolving Therapies

 

Targeting mTOR pathway alone using everolimus did not produce significant responses. An extended phase I study of the anti-IGF-1R monoclonal antibody cixutumumab with an mTOR inhibitor showed a partial but short-lived response. The use of the multikinase inhibitors sorafenib and sunitinib have also shown partial responses leading to a number of phase II studies whereas angiogenesis inhibitors have not been successful (http://www.clinicaltrials.gov). Other potential targets are antagonists of β-catenin and Wnt signaling pathway and SF-1 inverse agonists. The application of radionuclide therapy using 131I-metomidate has recently been explored. However, despite recent advances in dysregulated molecular pathways in ACCs, these findings have not yet been translated into meaningful clinical benefits. Lately immunotherapy (pembrolizumab) in phase II studies is under investigation.

FOLLOW-UP

 

Patients who have undergone an apparently curative resection should be followed up regularly using endocrine markers and abdominal imaging. After complete resection, radiological imaging every 3 months for 2 years and then every 3-6 months for a further 3 years is proposed. 18FDG-PET should be performed at regular intervals to detect recurrent disease at high risk patients. Patients on mitotane therapy should be regularly monitored measuring serum mitotane levels ensuring adequate replacement therapy. In case of recurrence not amenable to surgical excision patients should be enrolled in prospective clinical trials.

 

GUIDELINES

 

Fassnacht M, Dekkers O, Else T, Baudin E, Berruti A, de Krijger RR, Haak HR, Mihai R, Assie G, Terzolo M. European Society of Endocrinology Clinical Practice Guidelines on the Management of Adrenocortical Carcinoma in Adults, in collaboration with the European Network for the Study of Adrenal Tumors. Eur J Endocrinol. 2018 Jul 24.

 

Berruti A, Baudin E, Gelderblom H, Haak HR, Porpiglia F, Fassnacht M & Pentheroudakis G. Adrenal cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Annals of Oncology 2012 23 131-138.

REFERENCES

 

Petr EJ & Else T. Genetic predisposition to endocrine tumors: Diagnosis, surveillance and challenges in care. Semin Oncol 2016 43 582-590

 

Kassi E, Angelousi A, Zografos G, Kaltsas G, Chrousos GP. Current Issues in the Diagnosis and Management of Adrenocortical Carcinomas. In: De Groot LJ, Chrousos G, Dungan K, Feingold KR, Grossman A, Hershman JM, Koch C, Korbonits M, McLachlan R, New M, Purnell J, Rebar R, Singer F, Vinik A, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000-2016 Mar 6. PMID: 25905240

 

Chatzellis E, Kaltsas G. Adrenal Incidentalomas. In: De Groot LJ, Chrousos G, Dungan K, Feingold KR, Grossman A, Hershman JM, Koch C, Korbonits M, McLachlan R, New M, Purnell J, Rebar R, Singer F, Vinik A, editors. Endotext [Internet]. South Dartmouth (MA): DText.com, Inc.; 2000-2016 Feb 5. PMID: 25905250

 

Tacon L, Prichard R, Soon PSH, et al (2011) Current and emerging therapies for advanced adrenocortical carcinoma. The Oncologist 16:36-48

 

Fassnacht M, Kroiss M, Allolio B (2013). Update in adrenocortical carcinoma. J Clin Endocrinol Metab, 98:4551-4564

 

Tella SH, Kommalapati A, Yaturu S, Kebebew E. Predictors of survival in Adrenocortical Carcinoma: An analysis from the National Cancer Database (NCDB). J Clin Endocrinol Metab. 2018 Jun 21

Adrenal Suppression

CLINICAL RECOGNITION

 

Adrenal suppression, a form of secondary adrenal insufficiency (SAI), is a common clinical problem most often due to sudden cessation of chronic exposure to exogenous glucocorticoid administration or, rarely, after correction of endogenous hypercortisolism. It results from the inability of suprahypothalamic and hypothalamic centers of the hypothalamic-pituitary-adrenal (HPA) axis to recover their function and can last from days to months or years, depending on the dose and duration of the exposure to the glucocorticoid and patient’s idiosyncrasy. Exogenous glucocorticoids cause decreased secretion of corticotropin-releasing hormone (CRH) and other adrenocorticotropic hormone (ACTH) secretagogues, such as arginine-vasopressin (AVP) and alter the function of higher brain centers that regulate their secretion. Recovery of adrenal function may take as long as 1 to 2 years. In cases of endogenous hypercortisolemia adrenal suppression develops after the removal of a functional adrenal tumor secreting cortisol, or following successful removal of ACTH-secreting pituitary adenoma or other sources of ectopic ACTH secretion. Interestingly, the period to recover from adrenal suppression after the removal of ACTH-secreting pituitary adenoma caused by prolonged suppression of normal corticotrophs may be a predictor of sustained remission.

 

Regarding the definitions of adrenal insufficiency (AI), it is a disorder characterized by impaired adrenocortical function and decreased production mainly of glucocorticoids. Primary AI (PAI) is characterized, in addition by decreased production of mineralocorticoids (MCs) and/or adrenal androgens that occur in the setting of diseases affecting the adrenal cortex. Secondary AI (SAI) arises in diseases or conditions affecting the pituitary gland and the secretion of ACTH, while the affected hypothalamus resulting in abnormal secretion of corticotropin-releasing hormone (CRH) and other ACTH secretagogues defines the tertiary form of AI (TAI).

As adrenal suppression refers to decreases of cortisol secretion from the adrenal zona fascicularis the function of zona glomerulosa remains normal. Thus, hyponatremia is the main electrolytic disturbance observed, while circulating plasma potassium, renin, and aldosterone concentrations are within the normal range.

 

A broad range of severity can be seen as a result of complete or partial HPA axis suppression and concomitant adrenal gland atrophy. The true prevalence of overt adrenal insufficiency (AI) is probably rare as glucocorticoid treatment is gradually tapered before complete discontinuation leaving enough time for HPA axis recovery. However, due to the lack of specific symptoms the exact prevalence of AI following glucocorticoid tapering may be under-reported. Two recent systematic reviews reported that the percentage of patients with AI ranged from 0% to 100%, with a median (IQR) = 37.4% (13–63%), while when the studies were stratified by administration route, the percentages of patients with AI ranged from 4.2% for nasal administration (95% confidence interval [CI], 0.5–28.9) to 52.2% for intra-articular administration (95% CI, 40.5– 63.6); by disease, from 6.8% for asthma with inhalation glucocorticoids only (95% CI, 3.8 –12.0) to 60.0% for hematological malignancies (95% CI, 38.0 –78.6); by the dose from 2.4% (95% CI, 0.6 –9.3) (low dose) to 21.5% (95% CI, 12.0 –35.5) (high dose); by treatment duration from 1.4% (95% CI, 0.3–7.4) (less than 28 days) to 27.4% (95% CI, 17.7–39.8) (more than 1 year) in asthma patients.

 

The main symptoms of glucocorticoid insufficiency range from anorexia, fatigue, nausea, vomiting, dyspnea, fever, arthralgias, myalgias, and orthostatic hypotension to dizziness, fainting, and circulatory collapse. Hypoglycemia is occasionally observed in children and very thin adult individuals. Since 1-3% of adults worldwide are under long-term glucocorticoid therapy (Table 1), the awareness for adrenal suppression and the associated risk for glucocorticoid deficiency, as well as the appropriate treatment, are important clinical issues.

 

Table 1: Use of Glucocorticoid Therapy in Clinical Practice

Long-Standing Treatment
ENDOCRINE CAUSES
Replacement therapy Primary AI
Secondary AI
Adrenal suppression
Therapy
Congenital adrenal hyperplasia
Glucocorticoid resistance
Anti-inflammatory therapy Grave's opthalmopathy
NON-ENDOCRINE CAUSES
Immunosuppressive/
anti-inflammatory therapy
Rheumatic diseases- (lupus erythematosus, polyarteritis, rheumatoid arthritis, polymyalgia rheumatica)
Skin disorders- (dermatitis, pemphigus)
Other autoimmune diseases- (multiple sclerosis, myasthenia Gravis, vasculitis)
Hematological disorders- (lymphomas/ leukemias, hemolytic anemias, idiopathic thrombocytopenic purpura)
Gastrointestinal diseases- (inflammatory bowel
disease)
Liver diseases- (chronic active hepatitis)
Respiratory diseases- (angioedema, anaphylaxis, asthma, sarcoidosis, tuberculosis, obstructive airway disease).
Nephrotic syndrome
Suppression of host-versus-graft/graft-versus-host reaction- (bone marrow or organ transplantation)
Nervous disorders- (cerebral edema, raised intracranial pressure)
Acute Treatment
ENDOCRINE CAUSES
Suppression hypothalamic-pituitary-adrenal axis Cushing syndromediagnostic tests
NON-ENDOCRINE CAUSES
Several conditions Acute traumatic spinal cord injury
Post-operative additional therapy in severe neurological deficits even after surgery
Postoperative pain relief after severe bone operations
Fetuses between 24 and 34wk gestation (risk of preterm delivery)
Acute illness
or trauma
"Critical illness-related cortisol insufficiency"(CIRCI): vasopressor dependent septic shock and early severe
Acute Respiratory Distress Syndrome

AI: adrenal insufficiency

 

Many synthetic compounds with glucocorticoid activity have been developed in an attempt to maximize the beneficial and minimize the deleterious effects of glucocorticoids. The clinical efficacy of synthetic glucocorticoids depends on their pharmacokinetic, pharmacodynamic and molecular properties, which in turn determine the duration and intensity of glucocorticoid effects. According to their potency synthetic glucocorticoids are subdivided into short-, intermediate-, or long-acting. Treatment modifying factors, such as the age of the patient and the nature and severity of the underlying disease also influence synthetic glucocorticoid effects, duration, and doses administered.

 

The British National Formulary and the National Institute for Health and Care Excellence Clinical Knowledge Summary, both advise gradual glucocorticoid withdrawal in cases of patients that have received more than 40 mg prednisolone (or equivalent) daily for longer than one week; repeated glucocorticoid doses in the evening; glucocorticoids for more than three weeks; a short course of glucocorticoids within one year of stopping long-term glucocorticoid therapy; or have other risk factors for adrenal suppression.

 

PATHOPHYSIOLOGY

 

Supraphysiologic doses of glucocorticoids given even in small doses and/or for only a few days may result in considerable suppression of the HPA axis by decreasing CRH synthesis and secretion. The trophic and ACTH-releasing effects of CRH on pituitary corticotrophs are attenuated and the synthesis of propiomelanocortin (POMC), ACTH, and other peptides, are substantially decreased. In the absence of ACTH, the adrenal cortex temporarily loses the ability to produce cortisol, and when treatment with glucocorticoids is abruptly stopped transient glucocorticoid insufficiency ensues. It has been reported that the suppression of the HPA axis induced by exogenous glucocorticoids may persist for 6 to 12 months or rarely even longer, after treatment is withdrawn.

 

DIAGNOSIS and DIFFERENTIAL DIAGNOSIS

 

To support the diagnosis of adrenal suppression, several predictors of glucocorticoid-induced HPA axis hypofunction have been suggested the best being the duration and dosage of exogenous glucocorticoid administration (Table 2,3). A strong correlation has been found between prednisone maintenance doses above 5mg/d and a subnormal ACTH-stimulation test result. Hence, patients who are more likely to develop HPA axis suppression are those receiving high doses of glucocorticoids (>20-30mg hydrocortisone or equivalent) (Table 4) for a period longer than 3 weeks and patients who have developed overt Cushingoid features. In addition, the timing of drug administration may affect the degree of adrenal suppression. Thus, prednisolone in a dose of 5mg given at night before bedtime and 2.5mg in the morning will produce more marked HPA axis suppression compared to 2.5mg at night and 5mg in the morning. Higher evening doses block early morning ACTH surge whereas tissues sensitivity to glucocorticoids is increased in the evening and early night hours.

 

 

Table 2: Predictors of Glucocorticoid-Induced HPA Axis Suppression

Predictor Etiology/Risk of HPA Suppression
Type of steroid and potency Long-acting GCs lead to longer tissue life and longer suppression
Route of administration Systemic GC therapy (parenterally): increased risk
Timing
of administration
Decreased risk in alternate days scheme (from outset or converted before suppression);

Increased risk: different doses scheme during day:

Duration and cumulative dose Decreased risk in treatment ≤1week
Clinical features Patients with Cushing's Syndrome: increased risk

HPA: hypothalamic-pituitary-adrenal; GC: glucocorticoid.

 

Table 3: Examples of Different Glucocorticoid-Induced HPA Axis Suppression

HC/cortisone acetate: least potent/suppressive;

prednisone/prednisolone, methylprednisolone, triamcinolone: moderately suppressive;

dexamethasone: strongest suppression

Topical GCs: increased risk but infants at increased risk;
Inhaled GCs: increased risk versus oral/systemic GCs >risk children
Fluticasone proprionate (ciclesonide: recent drug, decreased risk);
Intraarticular GCs: transient suppression
Once-a-day dosing decreased risk intermediate/long acting GCs (prednisone/triamcinolone/dexamethasone);
Short-acting HC/ cortisone acetate: twice-a-day (at waking 2/3; 5PM 1/3 total daily dose); evening doses suppress normal early morning ACTH surge leading to increased suppression, treat with single morning dose
"Short-term" 14 days course systemic GCs decreased risk

ACTH: adrenocorticotropin; HC: hydrocortisone; GC: glucocorticoid

 

 

Table 4: Glucocorticoid Equivalent Dose Compared to Cortisol

equivalent dose (mg)
Short-acting, low potency
Cortisol 20
Cortisone 25
Intermediate-potency
Prednisolone 5-7.5
Methylprednisolone 4
Long-acting, high potency
Dexamethasone 0.75

 

Clinical awareness is crucial to identify patients with impending adrenal crisis. It is important to consider all patients with unexplained symptoms after glucocorticoid- withdrawal as candidates for possible AI and test them accordingly. An important feature that will raise suspicion of TAI (and SAI) besides drug history is the absence of skin pigmentation. Such patients have an intact renin-angiotensin-aldosterone system (RAAS) accounting for the differences in salt and water balance and clinical presentations compared to primary adrenal insufficiency.

 

Serum cortisol secretion at 08:00h if diagnostic tests are not feasible and until confirmatory testing is available can be considered a valuable screening method when AI is suspected. In patients with a low index of suspicion obtaining an 8AM cortisol and if the serum cortisol is > 15μg/dL, no further testing is needed. Similarly, a serum cortisol value <5 μg/dL suggests AI.

 

DIAGNOSTIC TESTS NEEDED TO DOCUMENT AI

 

Drug history and clinical features cannot be considered reliable tools for the evaluation of HPA axis function in patients treated with synthetic glucocorticoids. Several tests are commonly used in order to assess the degree of glucocorticoid-induced AI or HPA axis recovery (Table 5,6). Both the insulin tolerance test (ITT) and the metyrapone test have been employed as they are both highly sensitive. However, the risks involved with these tests do not justify their use compared to the rapid ACTH stimulation test or short synacthen test (SST) that can safely distinguish almost all cases of clinically significant adrenal suppression.

 

To evaluate the adequacy of HPA axis recovery, the SST is used to assess the capability of the adrenal cortex to respond to ACTH. However, because of the supraphysiologic ACTH levels achieved with the conventional SST (250 mcg of ACTH administered), if adrenal suppression is of recent onset, the adrenal gland may have not yet atrophied, and is still capable of responding to ACTH stimulation. In these cases, the low-dose SST (1 mcg of ACTH administered) has been proposed as an alternative as it results in lower plasma ACTH levels and thus less pronounced adrenal stimulation. It has recently been suggested that the low-dose SST is the best test to establish the diagnosis of SAI and TAI, whereas the high SST should be used for cases of primary AI. The use of salivary cortisol is also an effective alternative to serum cortisol when assessed in the high-dose ACTH test. Incremental cortisol response at the first SST was suggested as an important predictive factor of adrenal function recovery in SAI after exogenous glucocorticoid administration.

 

The CRH test can also be used in patients receiving glucocorticoids for prolonged periods, as it can assess both the ACTH and cortisol responses and can distinguish between SAI and TAI. In both conditions, cortisol concentrations are low at baseline and remain low after CRH administration. In patients with SAI, there is little or no ACTH response, whereas in patients with tertiary disease there is an exaggerated and prolonged response of ACTH, which is not followed by an appropriate cortisol response. On the contrary, patients with primary AI have high ACTH levels, which rise further following CRH while patients with hypothalamic disease show a steady rise in ACTH levels.

 

The prolonged ACTH stimulation test (depot or iv infusions 250µg cosyntropin over 8 hrs or over 24hrs) was suggested as a mean to differentiate between the different types of AI but is now rarely used in routine practice. In SAI or TAI, the adrenal glands display cortisol secretory capacity following prolonged stimulation with ACTH whereas in primary AI, they do not respond to ACTH being partially or completely destroyed.

 

In a recent systematic review of AI assessment after systemic glucocorticoid therapy, SST (conventional or low-dose) was the most frequently employed, but other tests were also used, including the insulin tolerance test (ITT, the “gold-standard”), the ACTH infusion, and the CRH tests.

 

Table 5: Diagnostic Tests Used to Diagnose Adrenal Insufficiency

Test / Sampling Cortisol Response
Short Synacthen test 250mg iv or im cosyntropin; samples at 0/30’/60’ Physiologic response:>500-550nmol/L (18-20µg/dL)
Low-Dose Synacthen
Test
1μg ACTH iv at
14:00: samples
10’ 15’ 20’ 25’
30’ 35’ 40’ 45’
Physiologic response:
>18 µg/dL (500nmol/L)
CRH stimulatory test
iv bolus 1 or 100µg/kg
or 100µgh-CRH/o-CRH
TAI: steady rise
in ACTH not followed by appropriate
cortisol response;SAI: no ACTH or cortisol response

ACTH: adrenocorticopic hormone; CRH: corticotropin-releasing hormone; im: intramuscular; iv: intravenous; PAI: primary adrenal insufficiency; SAI: secondary adrenal insufficiency; TAI: tertiary adrenal insufficiency

 

 

Table 6: Diagnostic Tests Not Commonly Used to Diagnose and Differentiate Adrenal Insufficiency

Test / Sampling Cortisol Response
Prolonged ACTH stimulation test
Depot or iv infusions 250µg cosyntropin over 8hrs(A): cortisol/24hr urinary cortisol/17OHCS before and after infusion or over 24hrs on 2(or3) consecutive days(B)
Physiologic response:
A:24hr urinary 17-OHCS
excretion increase 3-5-fold; serum cortisol>20μg/dL (550nmol/L) at 30’ and 60’; >25μg/dL (690 nmol/L) at 6-8hrs post-initiation infusion;B: at 4hrs >1000nmol/L (36μg/dL) beyond this time, no further increase; SAI: delayed response at 24 and 48hrs than 4hrs; PAI no response at either time
ITT
iv insulin (0.1-0.15U/kg); Samples 0 30’45’ 60’90’120’ with adequate clinical and biochemical hypoglycemia
Physiologic response:
>500nmol/L (18μg/dL)
overnight metyrapone test
30 mg/kg (max 3g)
at midnight; cortisol/ 11-deoxycortisol measured at 8.00h
the following morning
Physiologic response:
Increased ACTH plus peak 11-deoxycortisol >7 mg/dL.

ACTH: adrenocorticopic hormone; iv: intravenous; ITT: insulin tolerance test, PAI: primary adrenal insufficiency; SAI: secondary adrenal insufficiency; SST: short synacthen test; 17OHCS: 17-hydroxycorticoids, TAI: tertiary adrenal insufficiency

 

THERAPY

 

Glucocorticoid withdrawal is indicated when the use of the steroid is no longer needed or when significant side effects develop. The suggested method of glucocorticoid withdrawal is dose tapering to avoid the occurrence of AI.

 

Adrenal insufficiency is a potentially life-threatening medical emergency when presenting as adrenal crisis, which requires prompt treatment with hydrocortisone and fluid replacement. Once, clinically suspected, treatment should be initiated and not be delayed while waiting for definitive proof of diagnosis. Blood samples should be obtained for measurement of cortisol concentrations later, and the management approach should be similar to the resuscitation of any critically ill patient.

 

There is currently no consensus regarding rapid or slow tapering of glucocorticoids and exacerbation and/or relapse rates of the underlying diseases. The key action is that glucocorticoid withdrawal should not be abrupt. In clinical practice, patients being on any steroid dose for less than 2 weeks are not likely to develop adrenal suppression and are advised to stop therapy without tapering. The possible exception to this is the patient who receives frequent "short" steroid courses, as in asthma treatment. In longer regimens, the objective is to rapidly reduce the therapeutic dose to a physiologic level of cortisol (equivalent to 10-15 mg/ms/d) (Table 7). However, a recent systematic review of 73 studies demonstrated evidence of AI following low doses and short durations of glucocorticoid administration at less than 5 mg prednisolone equivalent dose/day, less than 4 weeks of exposure, cumulative dose less than 0.5 g, and following tapered withdrawal.

 

Table 7: Tapering After a Long-Term Glucocorticoid Regimen

1. Reduction by 2.5mg prednisolone or equivalent every 3-4 days over few weeks
2. Slower withdrawal until physiological level achieved (5-7.5mg of prednisolone)
3a. Decrease by 1mg/d prednisolone or equivalent every 2-4weeks (depending patient's general condition) until medication cessation
Or
3b. Switch to 20mg/d HC+ Decrease by 2.5mg/d every week until the dose: 10mg/d
4. After 2-3months on same dose SST or ITT
5a. Pass Response discontinuation of GC
Or
5b. No HPA axis recovery Treatment continuation+re-assessment

GC: glucocorticoid; ITT: insulin tolerance test; SST: short synacthen dose

 

Other tapering regimens suggest switching the patient to an alternate day administration of intermediate action glucocorticoids before cessation of treatment. Irrespectively of the tapering regimen used, if a glucocorticoid withdrawal syndrome, AI or exacerbation of the underlying disease develops, the dose being given at the specific time should be increased or maintained longer. Recent systematic reviews implied that the evidence for the tapering regimens used nowadays is not robust, despite the fact that rapid reduction to a physiologic glucocorticoid dose (5-7.5 mg prednisolone daily or equivalent), and the slow reduction thereafter, is the most frequently used regimen for clinicians.

 

Care should be given during the tapering regimens period on the interpretation of laboratory tests for cortisol levels measurement. The steroid dose before the test should be omitted (hold off evening and morning dose for hydrocortisone or prednisolone, longer for the other synthetic glucocorticoids); if serum cortisol secretion at 08:00h is > 15μg/dL, the tapering regimen changes to a rapid tapering off of exogenous glucocorticoids. Moreover, there are conditions that affect cortisol-binding globulin concentration (CBG) (↓: inflammation, nephrotic syndrome, liver disease, immediate postoperative period or requiring intensive care, rare genetic disorders; ↑: estrogen, pregnancy, mitotane). Systemic estrogens should be discontinued at least for 4 weeks prior to testing; estrogen patches are preferred since they do not affect CBG. Different criteria may apply according to the cortisol assay.

 

FOLLOW-UP

 

Since, there is evidence that AI may persist in 15% of patientsfor more than 3 years after glucocorticoid withdrawal, careful monitoring of patients and gradual glucocorticoid withdrawal should always be performed to avoid manifestations of adrenal suppression and/or an adrenal crisis or reactivation of the underlying disease. In general, plasma ACTH concentrations are not helpful in estimating the optimal glucocorticoid dose whereas mineralocorticoid replacement is not required.

All patients treated with glucocorticoids long-term should receive detailed instructions for glucocortiocoid supplementation equivalent to 100-150mg of hydrocortisone during major stresses (surgery, fractures, severe systemic infections, major burns) until their HPA axis fully recovers and to carry means of identification (medical alert bracelet).

 

Since full HPA axis recovery may take as long as one year or even longer, abrupt cessation of glucocorticoid treatment or quick tapering can precipitate an acute AI crisis. The diagnosis is a medical emergency, and treatment should be the immediate administration of fluids, electrolytes, glucose, and parenteral glucocorticoids.

 

GUIDELINE

 

Joint Formulary Committee, Glucocorticoid therapy. British National Formulary. London: BMJ Group and Pharmaceutical Press; 2013, 462.

 

National Institute for Health and Care Excellence.Corticosteroids—oral. http:// cks.nice.org.uk/corticosteroids-oral topic summary [accessed 15.09.20]

 

REFERENCES

 

Alexandraki KI, Kaltsas GA, Isidori AM, Storr HL, Afshar F, Sabin I, Akker SA, Chew SL, Drake WM, Monson JP, Besser GM, Grossman AB. Long-term remission and recurrence rates in Cushing's disease: predictive factors in a single-centre study. Eur J Endocrinol. 2013 Mar 20;168(4):639-48. doi: 10.1530/EJE-12-0921. Print 2013 Ap

 

Bansal P, Lila A, Goroshi M, Jadhav S, Lomte N, Thakkar K, Goel A, Shah A, Sankhe S, Goel N, Jaguste N, Bandgar T, Shah N. Duration of post-operative hypocortisolism predicts sustained remission after pituitary surgery for Cushing's disease. Endocr Connect. 2017 Nov;6(8):625-636.

 

Broersen LH, Pereira AM, Jørgensen JO, Dekkers OM. Adrenal Insufficiency in Corticosteroids Use: Systematic Review and Meta-Analysis. J Clin Endocrinol Metab. 2015 Jun;100(6):2171-80.

 

Joseph RM, Hunter AL, Ray DW, Dixon WG. Systemic glucocorticoid therapy and adrenal insufficiency in adults: A systematic review. Semin Arthritis Rheum. 2016 Aug;46(1):133-41.

 

Magnotti M, Shimshi M. Diagnosing adrenal insufficiency: which test is best--the 1-microg or the 250-microg cosyntropin stimulation test? Endocr Pract. 2008 Mar;14(2):233-8,

 

Neidert S, Schuetz P, Mueller B, Christ-Crain M. Dexamethasone suppression test predicts later development of an impaired adrenal function after a 14-day course of prednisone in healthy volunteers. Eur J Endocrinol. 2010 May;162(5):943-9.

 

Nicolaides NC, Chrousos GP, Charmandari E. Adrenal Insufficiency. In: De Groot LJ, Chrousos G, Dungan K, Feingold KR, Grossman A, Hershman JM, Koch C, Korbonits M, McLachlan R, New M, Purnell J, Rebar R, Singer F, Vinik A, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000-2017 Oct 14.

PMID: 25905309

 

Chrousos G, Pavlaki AN, Magiakou MA. Glucocorticoid Therapy and Adrenal Suppression. In: De Groot LJ, Chrousos G, Dungan K, Feingold KR, Grossman A, Hershman JM, Koch C, Korbonits M, McLachlan R, New M, Purnell J, Rebar R, Singer F, Vinik A, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000-2011 Jan 11. PMID: 25905379

 

Alexandraki KI, Grossman A. Adrenal Insufficiency. In: De Groot LJ, Chrousos G, Dungan K, Feingold KR, Grossman A, Hershman JM, Koch C, Korbonits M, McLachlan R, New M, Purnell J, Rebar R, Singer F, Vinik A, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000-2018 Aug 20. PMID: 25905345

Kidney Stone Emergencies

CLINICAL RECOGNITION

 

The acute passage of a kidney stone is the 9th most common cause of emergency room visits. Approximately 7-8% of women and 11-16% of men will have stone disease by age 70. The acute syndrome complex called renal colic implies obstruction of the collecting system or ureter, and the most common cause of obstruction is a kidney stone. Kidney stone colic is relatively constant in contrast to intestinal or biliary colic, which waxes and wanes or comes in waves. The onset of pain heralds the entrance of a stone into the collecting system and the ensuing obstruction. The intensity and location of the pain may vary with stone size, stone location, degree of luminal obstruction, and the suddenness of the obstruction but flank pain is very common. Referred genital pain is common with distal ureteral stones. Symptoms typically begin at night or the early morning hours with abrupt onset and awakening the patient from sleep. During the day, the onset of symptoms may follow heavy exercise and may be more gradual with an occasional prodrome of unilateral discomfort in the flank, testis or vulva on the side of the obstruction. The pain then becomes continuous, steady; and progressively more severe as it approaches a peak. For some, there are acute paroxysms of increasingly intense pain. Anorexia, nausea and vomiting commonly appear with the pain, and gross hematuria may be present. Overall, one-third of patients have a relatively rapid onset and reach peak pain in 30 minutes or less. Untreated, the pain may last for 4 to 12 hours, but most patients have presented to the emergency room by the time the pain becomes continuous, usually by two hours into the colic. Upon presentation, the pain is described as a 9 or 10 out of a scale of 1 to 10. Chills and fever may be present as well and should raise concern for infection as these symptoms are usually not present in uncomplicated urolithiasis. Similarly, hypotension also raises the likelihood of infection as the pain associated with renal colic typically induces hypertension and tachycardia.

 

PATHOPHYSIOLOGY

 

Stone-induced renal colic refers to an intraluminal cause, but non-stone related external compression of the ureter can induce the same symptom complex and be confused with the intraluminal presence of a stone. Renal colic can arise from three mechanisms: urinary obstruction, the most common cause, is due to a direct increase in intraluminal pressure and stretch of the nerve endings in the mucosa; local ureteral mucosal or collecting system irritation from direct contact of the stone; and interstitial edema and stretch of the renal capsule, particularly when there is a concomitant pyelonephritis. Stones are more likely to hang up and obstruct at naturally narrow regions of the upper urinary tract including the ureteropelvic junction, crossing of the iliac artery and vein, pelvic brim, and the ureterovesical junction.

DIAGNOSIS and DIFFERENTIAL

Diagnosis

 

The diagnosis is strongly suspected by the symptom complex. The examination reveals costovertebral angle tenderness with dysesthesia of the skin overlying the area along the flank, lower abdomen, groin, or genitalia. Gross or microscopic hematuria is present in 60% to 90% of patients with renal colic but is not required for the diagnosis.

 

Children with renal stones may present with more vague abdominal symptoms compared to the symptom complex in adults.  Therefore, abdominal pain in children and adolescents should call for a urologic evaluation if no diagnosis has been reached.

 

Differential Diagnosis

 

Acute renal colic may be caused by non-kidney stone events listed in Table 1.

 

Table 1. Causes of Acute Renal Colic

Intrinsic to the Collecting System
Kidney stones

Gross hematuria with clot formation

Tumor emboli

Renal papillary necrosis

Extrinsic to the Collecting System
Calyceal obstruction

Calyceal diverticula

Congenital ureteropelvic obstruction

Retroperitoneal fibrosis

Endometriosis

Dilation of the ovarian veins (pregnancy)

Mass lesions of the uterus

 

Acute onset of continuous, aching or dull pain that is non-colicky or flank pain without radiation to or toward the groin suggests a non-stone etiology. Common causes of acute non-colicky pain are listed in Table 2.

 

Table 2. Differential Diagnosis of Acute Non-colicky Renal Pain

Renal vein thrombosis

Pyelonephritis

Renal cortical abscess

Poststreptococcal glomerulonephritis

Rapidly progressive glomerulonephritis

Polycystic kidney disease

Medullary sponge kidney

 

DIAGNOSTIC TESTING

 

Imaging

 

The definitive diagnosis of acute renal colic relies upon radiographic imaging of the kidney and urinary tract to demonstrate the location, number, and size of the stones as well as the degree of obstruction. Non-contrast CT (NCCT) has become the imaging study of choice when evaluating patients with acute flank pain and suspected ureterolithiasis. It has both a high sensitivity and specificity for demonstrating the presence of stones and the ability to detect other abnormalities that maybe accounting for the symptoms. In addition, it has the advantage of providing information regarding stone number, location, size, and in some instances stone composition. It can also reveal signs of obstruction. The majority of patients evaluated by NCCT require no further imaging to determine the need for urological intervention. Many now advocate the use of low dose NCCT for the diagnosis of renal stones to reduce radiation exposure, particularly if the BMI is less than 30kg/m2.

 

Ultrasound is also a sensitive method for detecting ureteral stones in patients with renal colic and can be used as the initial imaging method in investigating these patients. However, the quality of ultrasound information is operator dependent and ultrasound has decreased diagnostic sensitivity. Kidney stones are common during pregnancy. Because fetal radiation exposure should be avoided, ultrasound is the primary radiologic procedure followed by MRI if necessary in pregnant women. NCCT should be used only in rare instances in pregnancy. In children ultrasound is the initial imaging procedure followed by low dose NCCT if needed.

 

A radiographic study done while the patient is in the emergency room will establish a definitive diagnosis, especially if it can exclude other causes of acute abdominal pain; will avoid a prolongation of the painful episode; avoid delay in treatment; and reduce the risk of loss of renal function when complete obstruction is present.

 

Laboratory Studies

 

The laboratory studies that should be obtained are shown in Table 3.

 

Table 3. Laboratory Studies

Complete Blood Count (CBC) Increased neutrophils may be due to a stress response or infection
Electrolytes
Creatinine Usually not markedly increased. A marked increase suggests solitary kidney, baseline kidney disease, or pre-renal injury due to dehydration
Calcium Hypercalcemia suggests the mechanism of stone formation and requires further evaluation
Uric acid Elevated uric acid levels suggest the mechanism for stone formation and requires further evaluation
Pregnancy testing in females of reproductive age
U/A Hematuria very common. WBCs if > 5/high powered field suggest infection
Urine culture and sensitivity if U/A abnormal or other signs of infection

 

Patients should be instructed to filter their urine in the hopes of retrieving a stone for analysis. Knowing the stone composition will help guide future preventive therapy.

TREATMENT

 

The goals of management during the acute phase of stone obstruction and renal colic includes: pain control and diagnostic procedures to determine the presence of a kidney stone in the collecting system and the extent of obstruction.

 

Pain management should be started soon after the patient arrives in the emergency room and should be continued until the episode has resolved. Nonsteroidal anti-inflammatory drugs (NSAIDS) (for example diclofenac, indomethacin or ibuprofen) are effective first line agents for acute pain treatment. If the pain persists or NSAIDS are contraindicated, narcotics, such as morphine sulfate 0.1 mg per kg body weight IM every four hours or meperidine (Demerol) 1.0 mg per kg body weight IM every three to four hours, may be used.  Intravenous lidocaine (1.5mg/kg) is another option that has been shown to be effective in reducing renal colic. Anti-emetic agents may be given along with the narcotics as nausea and emesis may occur with stone passage and commonly complicate narcotic use. If medical treatment is not sufficient consultation with urology and consideration of drainage or stone removal is indicated.

Alpha blockers, such as tamsulosin, may be used to facilitate the clearance of kidney stones. In a Cochrane review of 67 studies with 10,509 participants it was concluded that “alpha-blockers likely increase stone clearance but probably also slightly increase the risk of major adverse events (hypotension, syncope, palpitations, tachycardia). Subgroup analyses suggest that alpha-blockers may be less effective for smaller (5 mm or smaller) than for larger stones (greater than 5 mm)”. Smaller stones are more likely to spontaneously pass and therefore the advantages of alpha blockers are minimized but they may induce more rapid clearance. Additionally, alpha blockers also reduce renal colic.

 

The size of the stone is a major determinant of the need for surgical management vs. conservative management. Stones vary from less than 2 mm to greater than 2 cm in diameter. The majority of stones are less than 4 mm in width, small enough to pass spontaneously in most patients. A stone’s size is an important factor together with symptom severity, degree of obstruction, presence or absence of infection, and level of renal function in deciding whether to manage the stone initially by observation, awaiting spontaneous passage, or to intervene with a surgical procedure. Stones with a width of 5 mm or less have a 50% chance of spontaneous passage if in the proximal ureter and a better chance if in the distal ureter.  Overall, for stones ≤5 mm, approximately 68% will pass spontaneously. For stones >5 mm and ≤10 mm, an estimated 47% will pass spontaneously. One study found that stones > 9mm had only a 25% chance of spontaneous passage. Distal stones are more likely to clear than proximal stones (proximal ureter 48%, mid-ureter 60%, distal ureter 75% passage rate). Thus, in many patients with renal colic symptomatic treatment and close follow-up with the anticipation of stone passage is reasonable. The presence of infection, obstruction, refractory or difficult to treat pain, or deterioration of renal function indicates the need to urological consultation and the consideration of surgical intervention.

 

Urologic consultation should be obtained for possible surgical intervention for a number of reasons including stones with a low likelihood of spontaneous passage (large stones, proximal location), infection, obstruction, renal insufficiency or worsening renal function, and comorbidities that increase the risk of adverse outcomes (for example pregnancy). Depending upon the circumstances a number of procedures are available including ureteroscopic stone lithotripsy and extracorporeal shock wave lithotripsy for stone removal and percutaneous nephrostomy tube and JJ-stent for urinary drainage.

 

The presence of urinary tract infection increases the risk for development of pyelonephritis and/or pyonephrosis. Urgent intervention is therefore indicated, again regardless of stone size. Near-total or total ureteral obstruction predicts deterioration of renal function that may start within two weeks of presenting with stone disease and therefore indicates the need intervention.

 

FOLLOW-UP

 

Follow-up evaluation should be within one to two weeks of the acute event depending on the extent of intervention and whether there is risk for new obstruction from residual stones.  Metabolic evaluation using blood and urine tests may be performed after six weeks of recovery to guide specific preventative therapy. Stone analysis, and the results of urine and blood tests can guide decisions on preventive therapy. It should be recognized that after a first stone episode 30-50% of individuals have a recurrent stone within 10 years.

GUIDELINES

 

Pearle MS, Goldfarb DS, Assimos DG, Curhan G, Denu-Ciocca CJ, Matlaga BR, Monga M, Penniston KL, Preminger GM, Turk TM, White JR; American Urological Assocation. Medical management of kidney stones: AUA guideline. J Urol. 2014 Aug;192(2):316-24. PMID: 24857648

 

Türk C, Petřík A, Sarica K, Seitz C, Skolarikos A, Straub M, Knoll T. EAU Guidelines on Diagnosis and Conservative Management of Urolithiasis. Eur Urol. 2016 Mar;69(3):468-74.

PMID: 26318710

 

The EAU Recommendations in 2016. Medical Expulsive Therapy for Ureterolithiasis:

Türk C, Knoll T, Seitz C, Skolarikos A, Chapple C, McClinton S; European Association of Urology. Eur Urol. 2017 Apr;71(4):504-507. PMID: 27506951

 

REFERENCES

 

Favus M. Nephrolithiasis. In: De Groot LJ, Chrousos G, Dungan K, Feingold KR, Grossman A, Hershman JM, Koch C, Korbonits M, McLachlan R, New M, Purnell J, Rebar R, Singer F, Vinik A, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000- 2016 Dec 11.

PMID: 25905296

 

Gottlieb M, Long B, Koyfman A. The evaluation and management of urolithiasis in the ED: A review of the literature. Am J Emerg Med. 2018 Apr;36(4):699-706. PMID: 29321112

 

Fulgham PF, Assimos DG, Pearle MS, Preminger GM. Clinical effectiveness protocols for imaging in the management of ureteral calculous disease: AUA technology assessment.

J Urol. 2013 Apr;189(4):1203-13. PMID: 23085059

 

Campschroer T, Zhu X, Vernooij RW, Lock MT. Alpha-blockers as medical expulsive therapy for ureteral stones. Cochrane Database Syst Rev. 2018 Apr 5;4. PMID: 29620795

 

Jung H, Osther PJ. Acute management of stones: when to treat or not to treat? World J Urol. 2015 Feb;33(2):203-11. PMID: 24985553

The Postmenopausal Woman

ABSTRACT

The menopausal transition marks a time of great variability in reproductive hormones, and this variability can be responsible for specific symptoms, such as hot flashes and mood disturbances. Once a woman who is more than 45 years old has gone for 12 months without a menstrual period, she is considered to be menopausal and has consistently low circulating estradiol and elevated gonadotropins. Estrogen is the most efficacious therapy for bothersome vasomotor symptoms. Although estrogen exerts clear-cut protective effects on the cardiovascular system in premenopausal women, medical evidence does not support its use for the prevention of cardiovascular disease. Estrogen is generally not a first line agent for bone preservation in women without concurrent menopausal symptoms, despite its antiresorptive effects. Non-hormonal alternatives to estrogen and new, tissue specific estrogen complexes (TSECs) are now FDA approved and available for clinical use to treat common menopausal symptoms. For complete coverage of this and all related areas of Endocrinology, please visit our FREE on-line web-textbook, www.endotext.org.

 

INTRODUCTION

 

Menopause is associated with a constellation of physical changes.  Some of these changes are directly attributable to the loss of estrogen, including hot flashes, bone demineralization and vaginal dryness. Though a matter of controversy, an increased incidence of cardiovascular disease and dementia seem to be associated with both menopause and aging.  Furthermore, other conditions, such as breast, ovarian and endometrial cancer, are associated primarily with aging but certainly are impacted by ovarian hormones.

 

This review will address the menopausal transition, its common symptoms, and the risks and benefits of Hormone Therapy (HT), specifically, estrogen therapy and the selective estrogen receptor modulators (SERMs): raloxifene, tamoxifen and bazedoxifene, and other non-hormonal therapies.

 

DEFINITIONS

 

The Menopausal Transition

 

In 2001 [1] and again in 2012 [2], a Stages of Reproductive Aging Workshop (STRAW) was held to describe and define the various stages of the menopausal transition (Figure 1). On average, the menopausal transition lasts 4 years in duration and is divided into early and late phases. It begins when menstrual irregularity first appears, classically defined as either a “skipped” period or by an increase in variability of cycle length by more than 7 days. The menstrual irregularity that characterizes the menopausal transition occurs as the overall ovarian follicular complement decreases. However, the menstrual cycle and hormone changes of the early transition are best explained by a loss of the follicle cohort, rather than insufficient follicles to result in a single ovulation. This decrease in the available pool of growing follicles leads to a decrease in inhibin B production [3]. Reduced inhibin B removes the physiologic restraint on FSH that controls the process of folliculogenesis, and an increase in follicle-stimulating hormone (FSH) secretion is observed.  Early in the transition period, FSH levels are not consistently elevated, and may often vary considerably from month to month as the growing follicle cohort itself varies month to month. The follicular phase becomes notably shorter, and as a result, estradiol (E2) production is variable and even elevated at times. Follicle growth is more rapid, but ovulation may occur at smaller follicle diameters [4]. There is evidence that follicles may grow relatively rapidly in the preceding luteal phase, causing very short follicular phases, a phenomenon that has been named ‘luteal out of phase events’ [5]. The Study of Women’s Health Across the Nation (SWAN) collected daily urinary samples for an entire menstrual cycle annually from women at different stages of the menopause transition. A sharp drop-off in the proportion of ovulatory cycles begins at about 5 years before the final menstrual period (FMP) (Figure 2). Moreover, in cycles that appeared ovulatory, luteal progesterone production declines. Gonadotropins rise sharply beginning at 3 years prior to the FMP [6][7]. This early phase of the menopausal transition is associated with an increase in menopausal symptoms such as hot flashes, though initially, the increase may be relatively small as there may not necessarily be a reduction in the amount of circulating E2.  By the late transition, prolonged amenorrhea (defined as > 60 days) occurs, and is associated with a persistently reduced follicle pool and failure of folliculogenesis. At this point in the transition, estrogen deficiency begins to dominate, bone mineral density loss begins [8] and menopausal symptoms including hot flashes and vaginal dryness increase sharply in prevalence. Although the median duration of the transition is about 4 years, its duration is longer in women with onset at an earlier age, and can persist as long as 10 years or more in some cases [9].

Figure 1: The Stages of Reproductive Aging Workshop +10 staging system for reproductive aging in women [2].

PNG

Figure 2. Study of Women’s Health Across the Nation (SWAN).  Percent of cycles without evidence of luteal activity (ELA). [7]

 

Menopause

 

Menopause is defined as the cessation of menstruation for 12 months in a woman over age 45 and occurs at a median age of 52 years [10]. This event represents permanent failure of ovarian function secondary to depletion of the follicular pool. As such, supporting granulosa cells cease to produce estrogen and theca cells cease to produce androgens, and subsequently, ovarian estrogen and progesterone production stops. There is no established relationship between a woman's age at menarche and her age at menopause. However, it is well established that a woman's age at menopause is reflective of her mother's age at menopause [11]. Although few specific linked genes have been identified, there is heritability for age of menopause, and thus, several genes are likely involved in ovarian aging [12]. Menopause is known to occur approximately 1-2 years earlier in tobacco users [10].

 

Primary Ovarian Insufficiency

 

Primary ovarian insufficiency (POI), or premature ovarian failure (POF) has been defined as 3-6 months of amenorrhea accompanied by FSH levels greater than 40 IU/L on two separate occasions, at least one month apart in a woman less than 40 years old. POI is diagnosed in 5-10% of women who are evaluated for amenorrhea and the overall prevalence in the general population is thought to be around 1.1% [13]. The designation of “premature menopause” for such patients implies that menses will never happen again and this term should not be used. Rather, many recommend the use of the term “premature ovarian insufficiency (POI)” to describe the syndrome. POI and POF are more neutral terms, as young women with prolonged hypergonadotropic amenorrhea, unlike their older counterparts, are far more likely to have some intermittent ovarian function after the diagnosis has been made.

 

The treatment for POI usually consists of combined estrogen and progestin replacement. It is important to recognize that the risk to benefit equation of HT for women under age 40 who have ovarian failure differs from that for menopausal women aged 50-79. A preventive cardiovascular benefit for HT appears to be more likely in younger women.  Women with early loss of ovarian function are likely to spend more years of their lives exposed to the risk of bone demineralization, and therefore this important protective benefit of hormones is more likely to be realized. There are no current, evidence-based criteria to determine how to best provide hormone therapy to women with POI/POF, but it is widely assumed that hormone treatment up to the mean age at natural menopause should be considered in most cases, with a re-evaluation of risk to benefit once a woman attains the age associated with natural menopause in the population.

 

PHYSIOLOGIC CHANGES ASSOCIATED WITH AGING AND MENOPAUSE

 

Cardiovascular System

 

The largest health threat to women over age 50 is cardiovascular disease (CVD) [14]. In women age 45-49, the incidence of CVD is 3 times lower than men of matched age. However, data from the Framingham study have shown that by age 75-79, a woman's risk of heart disease increases and equals a man's risk for her age [15]. Women are less likely to be diagnosed correctly, less likely to undergo the correct revascularization procedure, and less likely to survive a major cardiac event than are men.  It is critical to develop new ways to identify preclinical disease amendable to intervention and prevention. Women appear to have risk factors that differ substantially from men, and include more social/emotional and autoimmune/inflammatory risks, along with more microvascular disease [16]. Further, vascular dysfunction is associated with more bothersome menopausal vasomotor symptoms [17]

 

Carotid intimal medial thickness (CIMT) has emerged as a strong predictor of subsequent disease and serves as a non-invasive marker of subclinical cardiovascular disease. El Khoudary, et al, have found associations between low endogenous SHBG and estradiol and elevated FSH with increased CIMT in perimenopausal women [18]. Endothelial function is also a predictor for CVD, and has been shown to decrease during the menopause transition. Due to these changes associated with diminishing ovarian function, researchers have been studying the use of hormone therapy to prevent the rise in CVD risk associated with menopause [19].

 

HT for the secondary prevention of coronary heart disease (CHD) was evaluated in The Heart and Estrogen/Progestin Replacement study (HERS) [20]. This trial included 2763 post-menopausal women with pre-existing CHD followed over 4 years. The objective of this study was to see if initiating HT would alter a woman's risk of future events. All participants were post-menopausal, younger than age 80 with a uterus and established CHD. Women were prescribed conjugated equine estrogen (CEE) 0.625 mg with medroxyprogesterone acetate (MPA) 2.5mg daily or placebo. The primary outcome was the occurrence of fatal or nonfatal myocardial infarction (MI). Secondary outcomes were other cardiovascular events: coronary revascularization, unstable angina, congestive heart failure, resuscitated cardiac arrest, stroke or transient ischemic attack and peripheral arterial disease. The results showed no significant differences in the occurrence of fatal or nonfatal myocardial infarctions by treatment. However, in the first year of the study, there were significantly more CHD events in the HT group, as well as a higher incidence of thromboembolic events (both deep venous thrombosis (DVT) and pulmonary embolus) and gallbladder disease when compared to placebo. The incidence of diabetes mellitus decreased by 3.5% over 4 years in the HT group. Similar to the results of the PEPI Study on intermediate cardiovascular markers [21], the HT group had a decrease in LDL cholesterol and an increase in HDL cholesterol when compared to placebo. These investigators concluded that HT did not reduce the risk of future cardiac events in post-menopausal women with established CHD. In addition, because of the increased incidence of adverse cardiac events in the first year of treatment, initiating HT in women with established CHD is not recommended. Based on the findings of the HERS Study, HT should not be initiated for secondary prevention of cardiovascular disease.

 

Studies have also focused on the possibility that use of HT during an optimal ‘window of opportunity’ in the early postmenopause could be effective for primary prevention of cardiovascular disease. The Women’s Health Initiative hormone therapy clinical trial did not find primary protection against CVD  in women treated for a mean of 5-7 years with either conjugated equine estrogen alone (women with a hysterectomy) [22] or estrogen plus the progestin, medroxyprogesterone acetate [23], and these results were consistent in cumulative 18-year follow-up [24]. When subgroup analyses were performed by age, women in the youngest age group (50-59 at enrollment), did not demonstrate significant benefit from HT. The Kronos Early Estrogen Prevention Study (KEEPS) tested the hypothesis that early intervention with estrogen delays the onset of atherosclerosis using a regimen of either conjugated equine estrogen or transdermal estradiol, both with cyclic administration of oral, micronized progesterone for 12 days each month. KEEPS did not demonstrate any between-group differences in CIMT or coronary calcium scores [25]. The rationale that hormones are protective of the vascular system when initiated early in menopause was supported, however, by the Early versus Late Intervention Trial with Estradiol (ELITE). The ELITE study found that oral estradiol treatment initiated within 6 years of menopause reduced CIMT compared to placebo, and this effect was not seen in women who initiated estrogen 10 or more years from menopause [26]. The long-term effect of time of HT initiation on CVD is therefore not established and HT is not currently recommended for primary prevention of CVD, regardless of age at initiation.  

 

LIPOPROTEIN CHANGES, CARDIOVASCULAR RISK, AND HT

 

The role of menopause in contributing to dyslipidemia has long been hypothesized. In women, total and low-density lipoprotein (LDL) cholesterol increase with age, and this increase is accelerated by menopause, whereas cardioprotective, high density lipoprotein (HDL) decreases. Moreover, the protective effect of HDL cholesterol appears to be diminished as women progress through menopause—possibly related to denser sub-particle size [27].  A rise in LDL has specifically been associated with the latter part of the menopausal transition and appears to be related to the loss of estrogen at this time of life [28]. In agreement with this finding is the relatively sharp upturn in CIMT observed in association with the late menopausal transition [29]. Through exercise, a low-fat diet, and cholesterol-lowering drugs, patients with high total and LDL cholesterol levels are able to significantly lower these lipoprotein levels and their subsequent risk for heart disease [30].

 

The Womens Health Initiative (WHI) trials describes a group of randomized, placebo controlled, clinical primary prevention trials that were designed to test the effects of HT, diet modification, and calcium and vitamin D supplements on CVD, fracture risk, and breast and colorectal cancer. The WHI had three overlapping clinical trials.  One was to test the effects of a low-fat diet on breast cancer and cardiovascular disease outcomes; one was to test the effect of calcium plus vitamin D on fracture outcomes, and one was to test the effects of hormone therapy in cardiovascular disease outcomes. The hormone therapy trial consisted of three study arms: The Estrogen + Progestin arm (conjugated equine estrogen (CEE) + medroxyprogesterone acetate (MPA) was administered to women with a uterus, the estrogen-alone arm (CEE) was administered to women without a uterus, and a placebo arm involved both women with or without a uterus. The WHI findings suggest that administration of HT does not protect the heart. While the initial analysis showed that CEE + MPA use was associated with a 24% overall increase in the risk of CHD (6 more heart attacks annually per 10,000 women using CEE + MPA) and an 81% increased risk of CHD in the first year alone after starting therapy, 18-year cumulative follow-up showed no difference in CHD and CVD-related mortality [24]. Women who had higher baseline LDL cholesterol levels at the beginning of the study were at particularly high risk of CHD with HT use [31]. Although the expected changes in lipoproteins were observed with hormone therapy (decreased LDL and increased HDL), there was no associated reduction in CHD risk.

 

The estrogen alone arm (CEE) differed from the CEE + MPA study in that it enrolled women who did not have a uterus, and who therefore did not need progestin. In this trial, 10,739 women with a prior hysterectomy, aged 50-79 years, were assigned to CEE 0.625 mg daily or to placebo. The study was stopped ahead of schedule in February 2004 for ‘futility’. During 7.1 years of follow up, estrogen provided no overall protection against heart attack or CAD in healthy post-menopausal women, most of whom were more than 10 years past menopause when they entered the study. In women 50-59 years of age at study entry, there was a suggestion of lower rates of heart attacks or procedures to revascularize thrombosed coronary arteries; however, these findings could be due to chance [22].

 

Data from the WHI estrogen-alone arm (CEE) supports the notion that coronary calcium accrual is prevented by early intervention with estrogen.  The WHI evaluated the presence of coronary artery calcium (CAC) burden to determine whether or not it differed based on treatment assignment. The WHI Coronary-Artery Calcium Study (WHI-CACS) evaluated 1,064 women aged 50 to 59 years after a mean of 7.4 years. CAC was evaluated by cardiac CT scans, which were performed blindly on patients to measure the CAC in these estrogen-alone participants. CAC scores were lower in women in the (CEE) alone group compared to those in the placebo group. The mean CAC score was 83.1 for (CEE) and 123.1 for placebo. After taking into account other heart disease risk factors, the risk of having mild-to-moderate CAC was 20-30% lower and the risk of severe CAC was 40% lower in the (CEE) group compared to placebo. After the trial ended, the calcium plaque build-up in the coronary arteries was lower in women randomized to estrogen compared to placebo [32].

 

In conclusion, studies show that most women have minimal CAC and minimal increases in carotid IMT prior to menopause. The findings imply strongly that ovarian hormones exert a protective effect on the cardiovascular system in premenopausal women, even though they do not appear to maintain a protective role after menopause. Despite these data, and secondary findings suggestive that early intervention with hormones may delay the onset of clinical heart disease, prescribing hormones for this purpose cannot be recommended based on the available data. These studies are unlikely to be the last word in this controversial field.  

COAGULATION

 

After menopause, there are noted changes in clotting parameters. There is an increase in procoagulation factors including fibrinogen, plasminogen activator inhibitor-1 (PAI-1), and factor VII, all of which cause a relatively hypercoagulable state. These increases are thought to be another contributor to the increase in cardiovascular and cerebrovascular disease in older women. With the administration of oral estrogen therapy, many procoagulation parameters improve, as evidenced by a decrease in fibrinogen and plasminogen levels; however, there is a higher risk for venous thromboembolism (VTE) due to increased liver metabolism of estrogen given orally [33].  

 

HT in currently used doses is associated with an approximately 3-fold increase in VTE events.  Transdermal estrogen preparations bypass liver metabolism and may be associated with the lowest VTE risk. Multiple observational studies have demonstrated fewer VTE and ischemic events with transdermal estrogen preparations compared to oral [34] [35-37]. SERM preparations also increase VTE risk. Tamoxifen increases VTE risk in a manner similar to oral estrogen, whereas raloxifene is associated with fewer VTE events than tamoxifen or estrogen [38, 39].  Bazedoxifene showed similar VTE risk compared to raloxifene in a randomized placebo-controlled trial [40]

 

Skeletal System

 

Osteoporosis is a major concern for postmenopausal women, leading to substantial morbidity and mortality. Fifty percent of women over age 65 have a compression fracture. Maintenance of bone mass is critical to prevent the development of osteoporosis. Height loss, up to several inches, and postural changes including kyphosis and lordosis are also caused by vertebral fractures. The mortality rate of women with hip fractures is 20% within the year following the fracture [41].

 

After peak bone mass is attained, usually around age 30, there is a slow, steady decline during the reproductive years, when approximately 0.7% of total bone is lost per year. At menopause, there is an accelerated rate of bone loss; 5% trabecular and 1.5% of total bone mass, on average, is lost per year. In the first 20 years after menopause, there is a 50% reduction in trabecular bone and 30% reduction in cortical bone, primarily due to the lack of estrogen [42].

 

Estrogen is responsible for promoting osteoblast (bone-forming cell) activity. It also inhibits bone remodeling and balances osteoblast and osteoclast (bone-resorbing cell) activity. As levels of serum estrogen decline in menopause, there is an increase in the rate of bone loss. As such, increased bone turnover increases serum calcium.  This increase in serum calcium, in turn, causes a decrease in parathyroid hormone (PTH) secretion, followed by calcinuria and decreased renal production of 1,25 dihydroxy-vitamin D. Vitamin D is responsible for intestinal calcium absorption and kidney tubular reabsorption. This domino effect causes a postmenopausal woman to lose 20 to 60 mg of calcium daily [43].

 

OSTEOPOROSIS SCREENING

 

It is a challenging public health problem to provide a cost-effective approach to identify women who are most likely to fracture, and to preferentially target them for screening and therapy.

 

An important and sensitive test to identify bone loss is a Dual Energy X Ray Absorptiometry (DEXA) scan. Usually two sites are analyzed-- the lumbar spine and the femoral neck (occasionally the radius is also checked). Scoring systems for evaluating Bone Mineral Density (BMD) are based on the T-score and Z-score. The T-score compares the patient's BMD to young women at peak bone mass whereas the Z-score compares the patient to women her own age. It is the T-score that is used to make a diagnosis.

 

The World Health Organization (WHO) has established the following definitions:

  1. normal BMD as a T-score => -1 standard deviation (SD) of the mean
  2. osteopenia as BMD between -1 and -2.5 SD
  3. osteoporosis as a T-score =< -2.5 SD

 

However, BMD via DEXA scan has a precision error of 2 to 6% depending on the site, which can amount to almost 1 t-score unit [44].

 

Bone density screening is useful, but does not provide all of the desired information about true fracture risk.  Low bone density alone will not cause a fracture, unless it is so low that activities of daily living cause bones to break.  Rather, women must have a combination of low bone density and a predisposition to falling that increases their risk. All major current guidelines state that BMD screening should begin at age 65 years for women of ‘average risk’ [45]. The rationale for waiting until age 65 to screen is that for most women, therapy will not need to be initiated before this time.  Most guidelines also agree that BMD screening can and should be used selectively for women younger than 65 years if they are postmenopausal and have other risk factors for fracture (Table 1).  Other considerations for BMD screening include estrogen deficient women of any age, vertebral anomalies and primary hyperparathyroidism.

 

Table 1. When to Screen for Bone Density Before Age 65 Years

Bone density should be screened in postmenopausal women younger than 65 years if any of the following risk factors are noted:

  •  Medical history of a fragility fracture
  •  Body weight less than 127lb
  •  Medical causes of bone loss (medications or diseases)
  •  Parental medical history of hip fracture
  •  Current smoker
  •  Alcoholism
    •  Premature ovarian failure
  •  Rheumatoid arthritis

 

In order to attempt to address the factors beyond bone density that can be used to predict fractures, the World Health Organization (WHO) developed the Fracture Risk Assessment Tool (FRAX) to identify those women who are at the greatest risk for fracture. FRAX was developed to calculate the 10-year probability of a hip fracture and the 10-year probability of a major osteoporotic fracture (defined as a clinical vertebral, hip, forearm or humerus fracture) taking into account femoral neck BMD and the risk factors listed below in Table 2. Clinicians can use the FRAX tool to make clinical decisions regarding BMD testing (http://www.shef.ac.uk/FRAX/index.aspx). FRAX can be used in women younger than 65 years to determine which women should have a BMD scan [46]. Those women with a FRAX 10-year risk of major osteoporotic fracture of 9.3% could justifiably be referred for DXA because that is the risk of fracture found in a 65-year-old Caucasian woman with no risk factors. It is important to note that FRAX does not provide data on fracture risk for women aged 40 or under.

 

While FRAX has been a highly utilized tool for clinicians, The American College of Physicians (ACP) recently suggested there is little evidence demonstrating effective treatment outcomes [47]. This limitation of the FRAX assessment was based on a randomized, controlled trial that showed that raloxifene significantly reduces clinical fractures in women ages 31-81 years but has similar efficacy regardless of a woman’s degree of  fracture risk [48]. Ultimately when using screening tools, it is important for clinicians to take into account not only the age, but also presence of risk factors when deciding whom to screen for BMD testing.

 

Table 2. WHO Technical Report: Fracture Risk Assessment Model

Risk Factors Included in the Fracture Risk Assessment (FRAX) Model
• Current age
• Rheumatoid arthritis
• Sex
• Secondary osteoporosis
• A prior osteoporotic fracture
• Parental history of hip fracture
• Femoral neck BMD
• Current smoking
• Low body mass index (kg/m2)
• Alcohol intake (3 or more drinks/day)
• Oral glucocorticoids ≥5 mg/d of prednisone for ≥ 3 month

 

AVOIDING BONE LOSS

 

Exercise, calcium and vitamin D supplementation can help protect women from bone loss. By engaging in regular weight-bearing exercise, women lose less bone than they would if they remained sedentary [49]. The Institute of Medicine recommends women ingest 1200 mg of dietary calcium and 400 IU dietary vitamin D daily to help protect from menopausal bone loss [50]. Supplementation with calcium and vitamin D if dietary levels cannot be achieved has been recommended.  However, concerns have been raised about calcium supplementation including increased risk of renal stones and cardiovascular events [51]. Data from several large clinical trials raise the possibility that a small but statistically significant risk for cardiovascular disease exists (Table 3). This risk does not seem to exist if a woman takes in calcium through dietary sources. It has been speculated that higher serum calcium levels are achieved with supplements but not when calcium is absorbed through consumption of calcium-rich foods, and that this transient; high circulating calcium can cause tissue calcification and dysfunction.

 

Table 3.  Calcium Supplementation and Risk of Heart Disease.

Author Study N Findings
Bostick [52] Iowa Women’s Health Study 34,486 Decreased risk        HR 0.66
Michaelsson [53] Swedish Cohort Study 61,433 >1400mg/day increased risk          HR 2.57
Chung [54] Meta-analysis 200 articles No association
Bolland [55, 56] WHI—CT ONLY 36,282 >1000mg/day Increased risk
Prentice [57] WHI—CT +OS >100,000 No association
Xiao [58] NIH-AARP diet and health study 388,229 No increased risk with supplements
Paik [59] Nurse’s Health Study 74,245 No increased risk with supplements
Donneyong [60] WHI – CT + Vit D 35,983 Heart failure reduced in women with highest risk for heart failure

 

Such cardiovascular risk has not been demonstrated with vitamin D. Two recent controlled trials did not demonstrate increased cardiovascular events with use of high-dose vitamin D supplementation [61, 62]. However, guidelines do not currently support daily supplementation with calcium or vitamin D for primary prevention of fracture in postmenopausal women [45]. Practically speaking, patients should be encouraged to eat as many calcium and vitamin-D rich foods as they can through their diet. Those who have documented vitamin D deficiency should be given supplements.

 

TREATING OSTEOPOROSIS

 

Treatment for osteopenia and osteoporosis includes weight-bearing exercise, dietary modification, assuring adequate calcium and vitamin D intake, and the introduction of other medications. There are several different types of medications that can be used to treat low BMD: bisphosphonates, SERMs, calcitonin, hormones, and denosumab are all clinically-proven anti-resorptives (Table 4).

 

Table 4. Treatments for Osteoporosis

Treatment and Prevention
Bisphosphonates
  • Alendronate (Fosamax) 10 mg daily tablet, 70 mg weekly tablet, or liquid formulation
  • Risedronate (Actonel) 5 mg daily tablet, 35 mg weekly tablet, or 150 mg monthly (75 mg tablet on 2 consecutive days)
  • Ibandronate (Boniva) 2.5 mg daily tablet, 150 mg monthly tablet, or 3 mg IV therapy every 3 months
  • Zoledronic Acid (Reclast) 5 mg IV therapy yearly
SERM 
  • Raloxifene HCl (Evista) 60 mg daily
Treatment Only
Calcitonin 
  • Calcitonin Salmon (Miacalcin or Fortical)  200 IU daily intranasal spray or 100 IU daily IM or SQ
PTH 
  • Recombinant PTH (1-34) Teriparatide (Forteo) 20 µg SQ daily
RANK-L ligand inhibitor
  • Denosumab (Prolia) 60mg SC q6months
Prevention Only
HT
  • Estrogen (see table 10 for detailed information)
TSEC
  • Conjugated estrogen + bazedoxifene (Duavee) 0.045mg/20mg daily tablet

 

Although most fractures occur in women with bone density in the osteopenic range, it is

not recommended to treat osteopenia without additional features that carry a more worrisome prognosis for fracture [63]. The approved medications for both treatment and prevention of osteoporosis include bisphosphonates and the SERM, raloxifene. Bisphosphonates have been a mainstay of therapy for many years, and act by inhibiting bone resorption. Although they have a long track record of efficacy and safety, prolonged and high-dose usage has been associated with the rare side effects of osteonecrosis of the jaw and atypical femoral fracture [63]. Recent research indicates that bone density is maintained for several years after discontinuation of treatment, and ‘drug holidays’ may help reduce the risk of developing adynamic bone. Recent guidelines recommend treatment for 5 years and do not recommend additional BMD assessment during this time. Bisphosphonates should be avoided in women of child-bearing potential as they deposit in the bone, have a very long half-life, and accumulate in fetal bone if they are given to the mother.

 

Raloxifene acts like a pro-estrogen on bone, lipids and liver and acts as an anti-estrogen on both the uterus and the breast. This makes its effects more favorable than tamoxifen, which acts like a mixed estrogen agonist on the uterus. The landmark MORE (Multiple Outcomes of Raloxifene Evaluation) trial evaluated the ability of raloxifene to prevent fractures in women with established osteoporosis. 7705 post-menopausal women were randomized to either 60 or 120 mg of raloxifene versus placebo. The risk of both vertebral and non-vertebral fractures was reduced in the groups treated with raloxifene, and BMD increased in both the hip and the spine in raloxifene treated patients [64]. Furthermore, a substantial decrease in the incidence of breast cancer was noted in raloxifene treated women, and the risk of having estrogen receptor positive invasive breast cancer was decreased when compared to placebo [65]. There was no difference between treatment groups with respect to the development of endometrial cancer.

 

For prevention of osteoporosis in postmenopausal or hypoestrogenic women, menopausal hormone therapy (when symptoms are present) or bazedoxifene/conjugated equine estrogens are appropriate agents [63]. A disadvantage of HT compared to bisphosphonates is the abrupt decrease in bone density that occurs when HT is stopped. Bazedoxifene is a SERM that has a similar profile to raloxifene, and thus, when combined with estrogen, appears to exert a neutral effect on the endometrium and can therefore be given without a concomitant progestin. This confers a significant advantage over HT for women with a uterus. This combination of SERM with estrogen, such as bazedoxifene with estrogen, is termed a tissue selective estrogen complex (TSEC). Bazedoxifene has a similar profile to raloxifene but has not yet been tested in a large clinical trial for outcomes related to breast cancer [66]. Thus far, clinical studies demonstrate no reports of breast concerns or benefits.  

 

Denosumab is a human monoclonal antibody to the receptor activator of nuclear factor-κB ligand (RANKL) that blocks its binding to RANK, inhibiting the development and activity of osteoclasts, decreasing bone resorption, and increasing bone density. This drug is approved for treatment, but not prevention, of osteoporosis. Denosumab can be given subcutaneously twice yearly to reduce the risk of vertebral, nonvertebral, and hip fractures in women with osteoporosis [67].

 

Parathyroid hormone (PTH) acts as an anabolic metabolite to stimulate bone production from osteoblasts, and is approved for treatment of osteoporosis. PTH decreases the incidence of new fractures and increases bone density. However, adverse side effects include hypercalcemia and gastrointestinal symptoms. Early rodent studies were concerning for possible bone tumor formation; however, post-marketing studies have not reported any cases. It remains that its approved use in humans is only for 24 months [68].

 

Calcitonin inhibits bone resorption, though not as effectively as other osteoporotic therapies. It is only available in intranasal or injectable forms as no effectiveness has been shown from oral formulations [69]. This is not generally considered first-line therapy but is a useful alternative when other medications are contraindicated.

Once a patient has been started on therapy, markers of bone turnover can be used to assess a patient's response. Urinary calcium, deoxypyridinoline, pyridinoline, hydroxyproline and N-telopeptides can be checked after 1-3 months of initiating treatment in selected cases [67]. DEXA scans, although they are currently the best method for determining BMD, should not be repeated too frequently since errors in interpretation of trends can occur and lead to inappropriate therapy [70]. It is recommended that DEXA scans be repeated no more frequently than every 2 years.

 

Central Nervous System

 

Vasomotor symptoms and “hot flashes” adversely affect the quality of life and functional status of most women during the menopausal transition. Hot flashes can occur in up to 85% of menopausal women. Col et al. estimated the duration of vasomotor symptoms in a longitudinal study on 438 women from the population-based Melbourne Women's Midlife Health Project. The onset and cessation of vasomotor symptoms were reported, and stratified according to whether or not HT was used. They found that the mean (SD) duration of bothersome menopausal symptoms for women who never used HT was 5.2 (3.8) years [71]. A meta-analysis of 35,445 women taken from 10 different studies appeared to confirm a median 4-year duration of hot flashes, with the most bothersome symptoms beginning about 1 year before the final menstrual period and declining thereafter [72]. However, two newer studies that have examined women longitudinally over a longer time frame indicate that the duration of vasomotor symptoms may be far longer than previously appreciated [73, 74].  These studies have found that hot flashes may last as long as 10 years in up to one quarter of women who report them. The earlier in life that they appear, the longer they may last, and among all racial/ethnic groups studied, African-American women appear particularly vulnerable to long duration, bothersome vasomotor symptoms.

 

The exact etiology of the hot flash has not been elucidated but a resetting and narrowing of the thermoregulatory system is believed to occur. In the past, hot flashes were thought to be related to a withdrawal of estrogen; however, there is no acute change in serum estradiol during a hot flash. Others have related hot flashes to variability in both estradiol and FSH.  It is thought that decreased estrogen levels may reduce serotonin levels and thus upregulate the 5-HT2A receptor in the hypothalamus. As such, additional serotonin is then released which can cause activation of the 5-HT2a receptor itself. This activation changes the set point for temperature and results in hot flashes [75]. More recent work has focused on the kisspeptin-neurokinin B-dynorphin neurons of the hypothalamus, the so-called KNDy neurons.  Ablation of the neurokin 3 receptor (NK3R) has been shown to abolish cutaneous vasodilatation in oophorectomized rats [76], and use of compounds that selectively block the NK3R have been shown to be effective in humans [77]. These exciting findings bring us closer to an understanding of the etiology of hot flashes and indicate the potential for novel treatments (discussed below).

 

MOOD

 

Significantly higher odds of depressive symptoms are reported by women who reach the late perimenopause. In the Study of Women’s Health Across the Nation (SWAN) [78], as well as 2 other longitudinal studies of the menopausal transition [79, 80], risk for depression was most pronounced in women who began the study with a low Center for Epidemiologic Studies Depression Scale score [78], indicating that the depressive symptoms were of new onset and appeared to be directly related to the menopausal transition. Follow-up studies using a Structured Clinical Interview for DSM-IV Axis I Disorders (SCID) confirmed that the late perimenopause is a vulnerable window for new-onset major depression [81].  The late perimenopause is also associated with a higher prevalence of sleep difficulty [82], which in turn is associated with depressive symptoms. Recent examination of anxiety symptoms in perimenopausal women indicate that, similar to depression, those with lower anxiety scores prior to the onset of the menopause transition are most vulnerable to a sudden escalation of anxiety and experience the greatest negative impact from their symptoms [83]. Not surprisingly, women with a lifetime history of anxiety and depressive symptoms during their menopausal transition report the lowest health related quality of life (HRQOL) [84], and poor sleep exacerbates these associations.

 

COGNITION

 

Women routinely complain of cognitive deficits around the time of menopause. Certain aspects of cognition appear to be related to a decline in estrogen, but many are simply related to the aging process itself. While some studies have demonstrated improved short term and verbal memory in postmenopausal women taking estrogen [85],others have not found such beneficial effects [86]. Greendale et. al. observed a sub-cohort of 2,362 SWAN participants longitudinally over 4 years to determine the effects of the menopausal transition and HT use on cognitive performance in midlife women. The outcomes analyzed were longitudinal performance in 3 separate areas: processing speed, verbal memory and working memory. The results of the study showed that, consistent with transitioning women's perceived memory difficulties, perimenopause was associated with a decrement in cognitive performance, characterized by women not being able to learn as well as they had during premenopause. Improvement rebounded to near-premenopausal levels once the transition was completed, suggesting that menopause transition-related cognitive difficulties may be time-limited. The initiation of HT prior to the final menstrual period had a beneficial effect, whereas initiation after the final menstrual period had a detrimental effect, on cognitive performance (68) [87]. More recently, the Cognitive Affective Study of the Kronos Early Estrogen Prevention Study (KEEPS-Cog) evaluated the impact of 4 years of HT on mood and cognition in early postmenopausal women. Various cognitive factors were not influenced by HT over 4 years, though a slightly positive effect on mood was observed in patients receiving oral conjugated equine estrogens. Mood and cognition did not differ between women receiving transdermal estrogen or placebo [88].

 

DEMENTIA AND ALZHEIMER’S DISEASE

 

The most common form of dementia is Alzheimer's disease (AD), which is 3 times more common in women than in men. Women with preexisting dementia or AD have been noted to have lower serum estradiol levels than women without dementia [89]. In observational studies, less AD has been observed in postmenopausal women who use estrogen and the effect was greater with increasing duration of use [90, 91]. In some trials, women with mild to moderate AD who were given estrogen had improvement in their dementia [92, 93], but this was not observed in all clinical trials [94, 95].  Estrogen has been believed to help prevent AD by regulating synapse formation in the hippocampus and by inducing acetycholinesterase and choline acetyltransferase, both of which are important in memory [96]. Estrogen may also improve cognitive function because of protection against neuronal toxicity caused by oxidation and increasing metabolism of serum amyloid P [97]. However, these molecular findings do not appear to translate into clinical benefits, as the WHI’s Mental Status (WHIMS) Trial demonstrated that hormone treatment with either (CCE+MPA) or (CCE) alone doubled the risk of AD and mild cognitive impairment.  These clinical trial findings do not support a long- term role of estrogen in the prevention or treatment of AD. However, considerable controversy remains, as the sensitivity of the testing used in the WHI may not have been adequate to detect early disease. As stated above, the KEEPS Trial did not note cognitive differences among women randomized to 2 types of estrogen plus progesterone or to placebo (86). This is noteworthy because KEEPS used a very detailed cognitive battery of tests.

 

LIBIDO

 

Loss of libido is a prevalent complaint in women of all ages and is present in approximately 9% of postmenopausal women [98]. Causes for a menopause-related decline in sexual interest may relate partly to a drop in both estrogen and testosterone with ovarian decline and aging, respectively. It is very important to consider the medication history and to screen for depression when clinically evaluating women with a complaint of diminished libido. In a survey of 35,381 women (the PRESIDE Study) [99], 10% reported decreased sexual desire; when women without concomitant depression or antidepressant medication were accounted for, the prevalence of desire disorder decreased to 6.3%.

 

Testosterone has long been considered as an agent that might promote libido in women.  Several well-conducted, double-blind, randomized trials of testosterone in menopausal women with decreased libido have demonstrated small, but clinically and statistically improved symptoms [100]. Testosterone has been used as a transdermal formulation in most of these studies and demonstrates efficacy with or without concurrent use of estrogen, in women with and without their ovaries. The APHRODITE study examined transdermal testosterone in 814 menopausal women over 52 weeks. Women were randomly assigned to receive either a patch delivering 150 or 300µg of testosterone per day or placebo.  Evaluation at week 24 demonstrated that the women on the 300µg testosterone patch noted a significantly greater increase in their 4-week frequency of satisfying sexual episodes in comparison to placebo, but this was not observed in the group receiving 150 µg per day. Both doses of testosterone patches were associated with significant increases in desire compared with placebo. Androgenic adverse events were greater in the group receiving 300 µg of testosterone per day. Breast cancer was diagnosed in 4 women who received testosterone (as compared with none who received placebo) [101]. The excess cases of breast cancer in women treated with testosterone may be due to chance. However, the possibility of a causal relationship must be considered as several published studies have shown that higher levels of endogenous testosterone and administration of exogenous testosterone are associated with the risk of breast cancer [102, 103]. Clearly, long-term data from large clinical trials using testosterone are lacking and are needed [100]. Of note, a recent trial of a testosterone gel for female libido was discontinued because of lack of efficacy.  There are no FDA-approved testosterone preparations available for women.

 

The only FDA approved medication for treatment of hyposexual desire disorder is flibanserin, marketed as Addyi. Flibanserin is a centrally-acting serotonin agonist/antagonist that increases female sexual desire and number of sexual acts [104]. These effects have been observed in the postmenopausal population, although it is not FDA-approved for this group of women. Adverse effects are generally mild and short-lived, except for a risk for hypotension and sedation if it is taken concurrently with alcohol, a problem that resulted in a black box warning and a need for prescribing clinicians to complete a risk evaluation and management strategy (REMS) certification before prescribing the drug [105].  Its effect size appears similar to that of testosterone (small, but statistically and probably clinically significant) [104].

 

Breast

 

After menopause and with aging, breast tissue is gradually replaced with increasing amounts of adipose tissue. This causes an age associated decrease in breast density, which makes mammography more effective in detecting breast disease. Breast cancer becomes more prevalent with advancing age with a lifetime risk of breast cancer in 1:8 women [106].

 

BREAST CANCER AND HT

 

Combined estrogen and progesterone treatment increase a woman's risk of developing breast cancer. The WHI trials demonstrated a detectably increased risk of developing invasive breast cancer after 3 years of combined HT use, with an unadjusted hazard ratio of 1.26 over 5.2 years of average follow-up [107]. Technically, the 95% confidence interval included 1.00, thus, the data could be considered ‘not significant’; however, this level of risk is biologically plausible, as it is similar to that seen in many observational studies, and similar to the small, incremental risk for breast cancer that is seen with later onset of menopause. The only risk factor identified in WHI patients for the development of invasive breast cancer was the duration of HT use. Patients taking hormones for 10 or more years were at greatest risk followed by patients using HT for 5 to 10 years. Women who took HT for less than 5 years had only a slight increase in risk. No correlation was noted between other risk factors--a patient's age, ethnicity, the 5-year Gail model risk score, body mass index (BMI), or family history--and the development of breast cancer. In women who had undergone hysterectomy and were randomized to CEE alone, no increase in breast cancer risk was observed; in fact, a decreased risk was observed in this group after 18-year follow-up [24, 108].

 

One of the ways in which HT might increase breast cancer is by increasing breast density. It has been noted that estrogen with cyclic micronized progesterone resulted in 16.4% more women with increased breast density [109].  A subset of 307 women in The Postmenopausal Estrogen/Progestin Interventions (PEPI) trial was studied to examine the effect of HT on mammograms. Of the group of women taking unopposed estrogen, 3.5% had an increase in breast density. Of the women taking both estrogen with progestin therapy, a 19.4-23.5% increase in breast density on mammography was noted, depending upon whether they took cyclic versus continuous MPA. Increased mammographic breast density is a strong independent risk factor (6-fold) for the development of breast cancer [110].

 

Case series and case-controls studies have suggested that patients taking HT who are diagnosed with breast cancer have a better prognosis than women not taking hormones, even when matched for stage of disease [111]. It has also been suggested that women who develop breast cancer while taking HT have their cancers detected at a more favorable stage and have less malignant disease [112]. These notions were disproven by the WHI Clinical Trial. Women randomized to combined HT with (MPA + CEE) had a higher risk of invasive breast cancer and mortality from breast cancer. Tumors in the women taking combined HT were comparable in histology and grade to the placebo group but were at a more advanced stage [107].

 

In contrast to combined E+P HT, E alone HT given to women without a uterus in the WHI, led to a decrease in breast cancer risk, which persisted after discontinuation of treatment and became statistically significant in the post-trial follow up study. After a median follow-up of 11.8 years, E alone treated women still had a lower incidence of invasive breast cancer (151 cases, 0·27% per year) compared with placebo (199 cases, 0·35% per year; HR 0·77, 95% CI 0·62—0·95; p=0·02 [113].

 

SCREENING FOR BREAST CANCER

 

The lifetime risk of developing breast cancer is 12%. Various organizations recommend breast cancer screening for average-risk women (Table 5). These guidelines all suggest an individualized approach with patients that includes consideration of a patient’s risk factors, as well as shared decision making based on a discussion of risks and benefits of screening. For average-risk women, mammography is the recommended screening modality.

 

Table 5. Breast Cancer Screening Guidelines*

ACOG[114] Offer annual or biennial mammogram starting age 40, start no later than age 50. Continue until age 75.
ACS[115] Offer annual mammogram starting at 40, start no later than age 45. Can offer biennial mammogram at age 55. Continue until within 10-years of life expectancy.
USPSTF[116] Biennial mammogram starting age 50. Continue until age 75.
*For average-risk females
ACOG = American College of Obstetricians and Gynecologists; ACS = American Cancer Society; USPSTF = United States Preventative Services Task Force

 

ASSESSING BREAST CANCER RISK

 

The Gail Model was developed to help clinicians determine if a patient was at higher risk than the general female population for the development of breast cancer [117].

The Gail Model takes into account the following characteristics:

 

  1. Age
  2. Age at menarche
  3. Age at first live birth
  4. Number of first degree relatives with breast cancer
  5. Number of previous breast biopsies
  6. Number of breast biopsies that were hyperplastic
  7. Race/ethnicity

 

This model provides an individualized risk for developing breast cancer over the next 5 years and over a lifetime. Other prospective scoring systems have been developed, but as of this writing there is no other dominant system that has proven to be superior to the Gail Model. By calculating a woman's risk of breast cancer with this model, a clinician can use the information to determine if a woman should consider chemoprophylaxis to reduce her risk of breast cancer. Note that the Gail model does not factor into account breast density or HT use. It also does not account for mutations, such as BRCA1 or 2, which have a profound effect on a woman’s risk of contracting breast cancer. Other risk factors not included are history of chest radiation prior to age 30 and extreme breast density.

CHEMOPREVENTION

 

In women who are considered high risk for breast cancer, chemoprevention therapies are approved to reduce breast cancer incidence. These therapies include SERMs (Tamoxifen and Raloxifene) as well as aromatase inhibitors (Exemastane and Anastrazole) [118].

 

Tamoxifen is indicated as adjuvant treatment for breast cancer. It is also prescribed for chemoprevention of breast cancer in high-risk women. Because tamoxifen is a SERM, it has both estrogenic and anti-estrogen actions. In the breast, it acts as an anti-estrogen. In the bone, on lipids and in the uterus, it acts like estrogen. Raloxifene is also a SERM, but has the advantage of acting as an anti-estrogen at the level of the uterus. Tamoxifen was found to be effective in breast cancer prevention in a trial that included 13,388 women who were at high risk for developing breast cancer because of 1) advancing age (>60 years old), 2) increased risk based on a Gail Model predicted risk of 1.66% over the next 5 years and age 35-59, or 3) a history of lobular carcinoma in situ. Women who were randomly assigned to tamoxifen experienced a 49% decrease in the incidence of invasive breast cancer compared to those who received a placebo. In addition, there was a decrease in the risk of estrogen receptor positive breast cancer and nodal involvement in those with breast cancer. Women randomized to tamoxifen also had fewer diagnoses of non-invasive breast cancer, such as ductal carcinoma in situ (DCIS) [119].

 

The STAR trial investigated the ability of tamoxifen compared to raloxifene in preventing  breast cancer in women at high risk for disease. All participants received either tamoxifen or raloxifene and took the drug for 5 years. In 2006, the results of STAR showed that both raloxifene and tamoxifen were equally effective in reducing breast cancer risk in post-menopausal women at increased risk of the disease. Women in the tamoxifen group and women in the raloxifene group had statistically equivalent numbers of invasive breast cancers (163 cases in 9,726 women in the tamoxifen group versus 167 cases in 9,745 women in the raloxifene group). Tamoxifen is known to be able to reduce breast cancer risk by 49%, and this study showed that raloxifene can also reduce breast cancer risk by half as well. As a result of this study, the FDA approved raloxifene as a second agent to help prevent invasive breast cancer in high-risk, post-menopausal women [120]. On an update of STAR trial, the risk ratio (RR; raloxifene: tamoxifen) for invasive breast cancer was 1.24 (95% confidence interval [CI], 1.051.47) and for noninvasive disease, 1.22 (95% CI, 0.951.59). Compared with initial results, the RRs widened for invasive and narrowed for noninvasive breast cancer. Toxicity RRs (raloxifene: tamoxifen) were 0.55 (95% CI, 0.360.83; P = 0.003) for endometrial cancer (this difference was not significant in the initial results), 0.19 (95% CI, 0.120.29) for uterine hyperplasia, and 0.75 (95% CI, 0.600.93) for thromboembolic events. There were no significant mortality differences [121].

 

To become active, tamoxifen must be metabolized by the hepatic cytochrome P450 enzyme system, specifically cytochrome P450 2D6 (CYP2D6), to its active metabolite, endoxifen.  Consequently, therapy with drugs that inhibit CYP2D6 may reduce the clinical benefit of tamoxifen by interfering with its bioactivation, particularly when these drugs are used for an extended period.  A significant percentage of patients with breast cancer experience a depressive disorder and are prescribed an anti-depressant, most commonly one in the selective serotonin reuptake inhibitor (SSRI) category. This is clinically relevant in the context of tamoxifen therapy, because SSRIs inhibit CYP2D6 to varying degrees.  Paroxetine is an irreversible inhibitor of CYP2D6, and therefore has the greatest potential to disrupt the biological activity of tamoxifen. A population-based cohort study was performed on 2430 women treated with tamoxifen and a single SSRI from 1993-2005. Of the group studied, 374 (15.4%) women died of breast cancer during follow-up. After adjustment for age, duration of tamoxifen treatment, and other potential confounders, absolute increases of 25%, 50%, and 75% in the proportion of time on tamoxifen with overlapping use of paroxetine were associated with 24%, 54%, and 91% increases in the risk of death from breast cancer, respectively (P<0.05 for each comparison). No such risk was seen with other anti-depressants [122].

 

The effectiveness of aromatase inhibitors for reduction of breast cancer incidence has also been demonstrated. The MAP3 trial investigated the incidence of invasive breast cancer with exemestane versus placebo in 5,560 high-risk postmenopausal women for up to 5 years [123]. Exemestane significantly reduced invasive breast cancer by 65% compared to placebo (95% CI, 0.18-0.70). There were no cardiovascular or thromboembolic side effects. However, follow-up study demonstrated worsened BMD after 2 years in the treatment group regardless of calcium and vitamin D supplementation [124]. Thus, for women receiving this therapy, close BMD screening is important. Anastrazole for prevention of breast cancer in high-risk postmenopausal women was studied in the IBIS-II trial [125]. One thousand nine hundred twenty women were randomized to anastrazole vs placebo for 5 years. A 53% reduction in invasive cancer was seen in the anastrazole group (95% CI, 0.32-0.68). Women who were concurrently treated with a bisphosphonate did not have significant bone loss, but the anastrazole-only group demonstrated worsened BMD after 3 years [126].

 

Thyroid Gland

 

As women age, the cumulative risk of hypothyroidism increases. Frequently, symptoms are ignored or misattributed to other causes, making the diagnosis difficult. It is recommended by ACOG that all women, even asymptomatic females, have a thyroid stimulating hormone (TSH) level measured beginning at age 50 years and every 5 years thereafter [127]. The American College of Physicians (ACP) also recommends periodic screening beginning at age 50 [128], while the American Thyroid Association (ATA) recommends that screening begin at age 35 [129].

 

Lower Reproductive Tract

 

The entire gynecologic tract contains estrogen receptors. As women become menopausal, the pelvic organs may be affected by the loss of estrogen resulting in vaginal atrophy, narrowing and shortening of the vagina and uterine prolapse, leading to high rates of dyspareunia. Furthermore, the urinary tract contains estrogen receptors in the urethra and bladder, and as the loss of estrogen becomes evident, patients may experience urinary incontinence (UI). Collectively, these symptoms, previously called vulvovaginal atrophy, have recently been renamed ‘genitourinary syndrome of menopause’ (GSM) [130]. While HT is effective in reversing changes associated with GSM [131, 132], it does not consistently help with symptoms of UI. The WHI Clinical Trial found that women who received HT and who were continent at baseline demonstrated an increase in the incidence of all types of UI at 1 year. The risk was highest for women in the CEE alone arm. Among women experiencing UI at baseline, the frequency of symptoms worsened in both arms and these women reported that UI limited their daily activities. This clinical trial evidence strongly suggests that HT should not be prescribed as part of a regimen for UI alone [133]. However, HT is highly effective in the treatment of vaginal dryness. Systemic or vaginal estrogen can be used for GSM, though locally applied estrogen is preferable if there are no systemic symptoms that need to be treated.  Very low doses can be used for this purpose. These low doses are believed to be safe for the uterus, even without concomitant use of a progestin. The data are currently insufficient to define the minimum effective dose, but vaginal rings, creams, and tablets have all been tested and demonstrated to reduce vaginal symptoms [134]. Ospemifene is a SERM that is FDA approved for the treatment of GSM symptoms [135].  It has a track record of endometrial safety [136] and in pre-clinical testing, was an effective antiresorptive agent for bone and may even have breast-protective effects [137]. These latter benefits remain to be proven in clinical trials. In 2016, prasterone, a formulation of dehydroepiandrosterone (DHEA), was FDA approved for the treatment of dyspareunia related to vulvar and vaginal atrophy. In a randomized controlled trial, 12-weeks of daily vaginal prasterone significantly alleviated dyspareunia compared to placebo [138]. The trial also demonstrated a significant drop in the vaginal pH, as well as improvement in vaginal dryness.

 

Adrenal Gland

 

The adrenal gland is responsible for producing androstenedione, dehydroepiandrosterone sulfate (DHEA-S) and, indirectly, total testosterone. After the menopausal years, androstenedione levels decrease by 62%, DHEA-S levels decline by 74% and testosterone, produced by the peripheral conversion of androstenedione, decreases by up to 25%. Circulating estrone, which is produced from the peripheral conversion of androstenedione, increases after menopause, whereas estradiol, which is produced from the peripheral conversion of estrone, declines.  The menopause-associated drop in estrogen is related to a significant decline in sex hormone binding globulin (SHBG), resulting in a higher free testosterone level [139]. This increase in free androgens may be responsible for the clinical problem of increased facial hair and androgenetic alopecia that accompanies the postmenopausal years for some women.

MENOPAUSAL TREATMENT

Figure 3: The Hormone Health Network has developed a self-administered algorithm for menopausal women to help them determine whether or not hormone therapy is a reasonable option for them. http://www.hormone.org/MenopauseMap.

 

Non-Hormonal Treatment

 

SELECTIVE SEROTONIN REUPTAKE INHIBITORS (SSRIs)

 

When HT is contraindicated, (i.e., history of breast cancer), women with hot flashes may be treated with non-hormonal prescription drugs; one such class is the SSRIs [140, 141]. Once initiated, the relief of vasomotor symptoms usually occurs within a week, more rapidly than the relief of depressive symptoms, which usually takes 6 weeks or longer. The most common side effects of these drugs are nausea and sexual dysfunction but use of the lowest dose may minimize these effects.

 

Though not as drastic of a reduction when compared to HT, the SSRIs result in a modest improvement in symptoms. A long-acting mesylate salt of paroxetine, 7.5mg, has been FDA-approved to treat hot flashes [142]. Non-approved SSRIs that have been tested and have clinical efficacy include paroxetine (non-mesylate), escitalopram, citalopram, fluoxetine and sertraline [141].

 

SEROTONIN-NOREPINEPHRINE REUPTAKE INHIBITORS (SNRIS)

 

Venlafaxine is a combined serotonin and norepinephrine reuptake inhibitor that has shown promise in reducing the severity of hot flashes in symptomatic women. A randomized trial was conducted in 229 women for 4 weeks where women with breast cancer received either varying doses of venlafaxine (37.5, 75 or 150 mg/day) versus placebo. There was a significant reduction in hot flashes in women receiving all doses of venlafaxine in comparison to placebo.  Common side effects included nausea or vomiting, which are usually limited to the first 1 to 2 weeks of treatment. Other side effects include lethargy, dizziness, constipation and sexual dysfunction [143].

 

GABAPENTIN

 

A randomized, double-blind, placebo-controlled trial was conducted on 197 women aged 45-65 years, who were menopausal and having at least 14 hot flashes per week. These women were randomized to receive either gabapentin 900 mg daily or placebo for 4 weeks. Of women assigned to receive gabapentin, hot flash scores decreased by 51% as compared with a 26% reduction in the placebo group, from baseline to week 4. These women reported greater dizziness, unsteadiness and drowsiness at week 1 compared with those taking placebo; however, these symptoms improved by week 2 and returned to baseline levels by week 4 [144]. A 2009 meta-analysis confirmed consistency across several clinical studies [145]. The dose range of gabapentin is broad, and although many clinical trials use doses of 900 mg, less may work well for individual patients.  The chief limiting side effects of gabapentin are drowsiness, dizziness (which can present a hazard for falls), and weight gain.

 

NEUROKININ B RECEPTOR (NK3R) INHIBITORS

 

Neurokinin B acting on its receptor, NK3R, at the level of the hypothalamus induces vasomotor symptoms typical of menopause. It is hypothesized that variable expression of NK3-R and interaction with its ligand is responsible for the differences in reported hot flashes experienced by menopausal women. The TACR3 gene codes for NK3-R. Genome-wide association studies performed on 17,695 women from the WHI trial and observational studies demonstrated significant genetic variation in TACR3 in women who reported vasomotor symptoms [146]. An oral NK3R antagonist completed phase II clinical trials and demonstrated a 45% decrease in the number of hot flashes per week as compared with placebo [147]. This drug is not associated with the side-effects of estrogen therapy, and further study will determine its efficacy and safety for use.

 

NON-PHARMACOLOGIC

 

Non-pharmacologic options for treatment of menopausal symptoms have yet to show proven benefit in large clinical trials. There are mixed results from trials evaluating the benefits of acupuncture for treatment of menopausal symptoms including vasomotor symptoms, insomnia and mood. As acupuncture is a generally low-risk therapy, it is at the discretion of the patient to pursue this treatment modality, but effectiveness in large trials is lacking. Phytoestrogens, which are estrogen-like compounds found in products such as soy, have no proven benefit in treatment of menopausal symptoms. Chinese herbal remedies likewise have not been shown to significantly alleviate symptoms, with or without acupuncture. The MsFLASH trial is a randomized controlled trial that showed reduction of insomnia in peri- and post-menopausal women with hot flashes who were treated with cognitive behavioral therapy for insomnia (CBT-I) compared to menopause education control [148]. Women who practiced CBT-I had significant reduction of insomnia after the 8-week intervention, and these results persisted at 24-week follow-up despite having no effect on daily hot flash frequency. Practical daily lifestyle habits including exercise, dressing in layers, consuming cold drinks, avoiding caffeine and alcohol may help alleviate symptoms.  

 

Hormonal Treatment

 

HT is utilized by many women for treatment of bothersome menopausal symptoms. As outlined above, there are specific risks and benefits associated with HT that may not make it suitable for some women.  Moreover, many women have a tendency to shun HT because the level of discourse about its true benefits and risks are so fraught with drama! It is important for the menopause care provider to be knowledgeable about the benefits and potential risks of hormonal therapies and to have some facility with non-hormonal alternatives. This approach allows the clinician to engage the patient in truly shared decision making. It is important to maintain clear lines of communication with menopausal patients who are struggling with bothersome symptoms, because their subjective improvement is frequently the sole arbiter of success of treatment, and it is what all risks must be balanced against.

 

HT is the most effective treatment for vasomotor symptoms and vaginal dryness caused by the loss of endogenous estrogen production. In addition, it acts like an anti-resorptive and is therefore osteoprotective and also has been shown to reduce the incidence of colon cancer by almost 40%. As mentioned earlier in this review, it is well established that HT changes the lipoprotein profile favorably, although these latter changes do not translate into reduced cardiovascular morbidity.

 

However, unopposed estrogen use in women who have a uterus creates a risk for developing endometrial hyperplasia and cancer. Therefore, estrogen replacement must be accompanied by a progestin. In patients with a uterus who were given estrogen alone in The Postmenopausal Estrogen/Progestin Intervention (PEPI) Trial, 62% developed endometrial hyperplasia over 3 years. By identifying this pathology early, patients were medically treated with high doses of progestins so that no patients developed endometrial cancer [21]. It is the standard of care to give women estrogen with a progestin when they have a uterus.

 

The decision to prescribe HT must be based on each individual patient, taking into account the risk factors involved and creating a favorable benefit to risk ratio. To date, acceptable reasons to prescribe HT include relief of severe vasomotor symptoms and to address GSM. There is sufficient medical evidence to consider a trial of HT for women with adverse mood or sleep symptoms in association with their menopause [149]. At present, there is no indication for using HT for the prevention of cardiovascular disease, dementia/AD, or osteoporosis, or for the prevention of colon cancer, as the risks outweigh any potential benefits, although as mentioned earlier in this review, there are suggestions that premenopausal HT may have protective effects in some cases.

A key factor in the decision tree for the initiation of HT is the individual risk of breast cancer, which is a real and serious concern. It is contraindicated to prescribe HT to patients with a history of breast cancer and it is not recommended to give HT to those with a high-risk profile. The adverse events demonstrated in patients taking combined estrogen-progestin HT included a 26% increase of invasive breast cancer, with the excess risk starting to be observed after 3 years of combined HT use.  It is important to note that estrogen alone treatment of women without a uterus did not increase the risk of breast cancer.

 

Recommendations for prescribing HT should be based upon the randomized, clinical trial results of the WHI, as highlighted throughout this review, as they currently constitute the best available medical evidence. Although the WHI studied the Prempro® formulation only, it is biologically plausible that other systemic formulations, including the transdermal patch, will carry similar risks and benefits and it should not be assumed that switching HT formulations protects a patient from adverse events.

 

However, The Estrogen and Thromboembolism Risk study, a multicenter case-control study of thromboembolism among postmenopausal women aged 45-70 years, demonstrated an odds ratio for venous thromboembolism in users of oral and transdermal estrogen to be 4.2 (95% CI, 1.5-11.6) and 0.9 (95% CI, 0.4-2.1), respectively, when compared with nonusers[34]. This has led ACOG, NAMS and the Endocrine Society to recommend that clinicians take into consideration the possible thrombosis-sparing properties of transdermal forms of estrogen therapy [140, 141, 150].

Women with vasomotor symptoms may consider short-term HT use at the lowest effective dose. Women who are currently taking HT and are asymptomatic, should be encouraged to periodically discontinue HT use to see whether or not symptoms return. Finally, women who desire long-term HT use for quality of life reasons (after appropriate counseling) should be evaluated regularly and their decision to continue HT periodically reassessed.  

HT REGIMENS: CONTINUOUS COMBINED AND CYCLIC REGIMENS

 

There are many ways to prescribe HT: oral tablets, patches, creams, sprays (Table 6). Considering the importance of including a progestin, there are several different modalities of administering these medications as well. This includes continuous combined and cyclical administrations. The continuous combined formulation administers both the estrogen and progestin hormones every day. Cyclical administration means that hormones are given in a cycle: 1) unopposed estrogen is given continuously 2) progestin is added. This regimen can be a cycle every 3 days (e.g. Ortho Prefest), every 14 days (e.g. Premphase), or at the discretion of the prescribing physician (e.g. every 3 months). Although generally believed to be safe, if progestins are given less frequently than monthly, the potential for hyperplasia exists and endometrial monitoring should be considered [151].

 

In women just entering menopause, the cyclical administration of the estrogen and progestin is usually the simplest choice. These patients can easily make the transition from taking a low dose oral contraceptive pill in the menopausal transition (frequently prescribed to control the irregular vaginal bleeding during that time) to the cyclical form of HT. At the onset of HT, most women will experience a withdrawal bleed at the end of the treatment month. Gradually, as the endometrium thins and becomes atrophic, some women will become amenorrheic on this regimen. Although irregular vaginal bleeding is uncommon, any abnormal uterine bleeding should be investigated. Another advantage of cyclical administration is that women will know when to expect bleeding.

 

Advantages of giving continuous combined therapy is that a lower dose of progestin can be used and patients should not expect a withdrawal flow at the end of the treatment month. Eventually, most women become amenorrheic on this regimen. Some women also develop irregular and inconvenient vaginal spotting or bleeding. This most frequently occurs in women who have recently entered menopause and still have an endometrial lining.

 

Besides oral preparations, HT can be administered in a variety of other ways. Estrogen can be delivered through a vaginal ring that delivers either 0.05 or 0.1 mg/day of estradiol acetate over a three-month period. It may also be given transdermally as 17β-estradiol with norethindrone acetate or levonorgestrel. Progesterone can be administered through a levonorgestrel-releasing IUD which can be left in place for up to10 years. Finally, vaginal preparations of progesterone are also available. More recently, transdermal estradiol sprays and gels have been FDA approved (Evamist ®, Divigel, and Elestrin).  These preparations are relatively short acting and sometimes need to be used more than once a day. All are FDA approved for the treatment of hot flashes.

 

Table 6. HT FORMULATIONS

Trade Name    Estrogen        Progestin       Dose
Vasomotor Symptom Therapies
Premarin Conjugated Estrogen - 0.3 to 1.25 mg PO daily
Cenestin Synthetic Conjugated Estrogen - 0.3 to 1.25 mg PO daily
Menest Esterified Estrogen - 0.3 to 1.25 mg PO daily
Estrace 17 β-estradiol - 1-2 mg PO daily
Estinyl Ethinyl estradiol - 0.02 to 0.05 mg PO 1-3 x daily
Evamist 17 β-estradiol - 1-3 sprays daily
Alora, Climara, Esclim, Menostar, Vivelle, Vivelle Dot, Estraderm 17 β-estradiol - 1 patch weekly-twice weekly
Estrogel 17 β-estradiol   - 1.25 g daily transdermal gel (equivalent 0.75 mg estradiol)
Estrasorb 17 β-estradiol - 2 foil pouches daily of transdermal topical emulsion   
Activella Estradiol 1 mg  Norethindrone Acetate 0.5mg  1tab PO daily
FemHRT Ethinyl Estradiol 5 mcg Norethindrone Acetate 1 mg  1tab PO daily
Ortho Prefest 17 β-estradiol 1 mg  Norgestimate 0.09 mg  First 3 tablets contain estrogen, next 3 contain both hormones; alternate pills every 3 days
Premphase Conjugated Estrogen 0.625 mg  Medroxyprogesterone Acetate 5 mg  First 14 tablets contain estrogen only and remaining 14 tablets contain both hormones.

1tab PO daily

Prempro Conjugated Estrogen 0.625 mg  Medroxyprogesterone Acetate 2.5 or 5 mg  1tab PO daily
Combipatch 17 β-estradiol Norethindrone acetate 1 patch transdermal twice weekly
Climara-Pro 17 β-estradiol Levonorgestrel 1 patch weekly
Angeliq 17 β-estradiol Drosperinone 1tab PO daily
Genitourinary Symptom Therapies
Estrace 17 β-estradiol vaginal cream - 2-4 g daily x 1 week, then 1 g three times weekly
Premarin 17 β-estradiol vaginal cream - 0.5 g daily for 21 days on, 7 days off or twice weekly
Vagifem 17 β-estradiol vaginal tablet - 10 mcg per vagina daily x 2 weeks, then 2 times per week
Estring Estradiol vaginal ring - 1 ring inserted vaginally every 3 months
Duavee Bazedoxifene 20mg Conjugated equine estrogen 0.45mg - 20/0.45mg daily
Ospemiphene - - 60mg PO daily
Prasterone - - DHEA 6.5mg inserted vaginally daily

 

TSECs—TISSUE SPECIFIC ESTROGEN COMPLEXES

 

The combination of bazedoxifene/conjugated equine estrogens represents yet another novel approach to hormone therapy. The combination of bazedoxifene, a SERM, with estrogen allows the clinician to apply estrogen where it is most beneficial—reducing or eliminating hot flashes, while the SERM bazedoxifene exerts anti-estrogenic effects at the target tissues where estrogen action is unwelcome—the endometrium and the breast [66]. Thus, the combination of bazedoxifene and conjugated equine estrogens is effective as an antiresorptive agent in bone and does not cause endometrial stimulation. With the bazedoxifene/conjugated equine estrogen combination, the clinician can avoid having to give progestin and avoid irregular or breakthrough bleeding.

 

SUMMARY

 

In conclusion, this review has highlighted the major health concerns faced by the post-menopausal woman. Cardiovascular disease becomes more prevalent with the loss of estrogen and the decrease in endothelial function and HDL cholesterol levels that occur concurrent with menopause. Osteoporosis is another serious potential problem that the aging woman faces and can be prevented by careful screening and early treatment. Cognitive decline and memory changes occur as aging ensues and AD becomes more prevalent, making it more difficult for aging women to maintain an independent lifestyle. Finally, breast cancer becomes more prevalent with advancing age. The increased risk of breast cancer needs to be considered when choosing a treatment plan for the post-menopausal woman.

 

There are a variety of treatments available to protect women from developing serious health problems. First and foremost, a healthy lifestyle is the best preventive medicine. HT will control a patient's vasomotor symptoms, prevent bone loss, maintain a favorable lipoprotein profile, and help prevent vaginal and urogenital atrophy. Other benefits of HT include the reduction in the incidence of colon cancer. The SERM, raloxifene, also can be used to treat osteoporosis in menopausal women. The advantage of a SERM compared to HT is its lack of endometrial stimulation and reduction in the risk of breast cancer. The prevention of bone loss and the beneficial effects on lipoprotein levels with SERMs are similar to those seen with HT.

 

The role of HT has changed over the years as its risks and benefits have been clarified through carefully designed randomized trials, most notably, the WHI. For a low-risk woman with moderate to severe vasomotor symptoms, the introduction of HT is an effective option and patients will improve. However, the clinician needs to evaluate each patient independently and take into account the individual risk profile, including family history, in order to determine which form of treatment is most appropriate. The ability to modulate estrogen action via the development of SERMs provides the hope that a 'perfect' SERM can be produced, which will relieve vasomotor symptoms, protect the bone and the heart, maintain a favorable lipoprotein profile, and be anti-estrogenic to the endometrium and the breast. Until then, non-hormonal alternatives are available for women who cannot or do not wish to take HT.  Prudent clinical judgment and an individualized assessment of risks and benefits for patients using the currently available medical evidence remains the most appropriate approach.

 

REFERENCES

 

  1. Soules, M.R., et al., Executive summary: Stages of Reproductive Aging Workshop (STRAW). Fertil Steril, 2001. 76(5): p. 874-8.
  2. Harlow, S.D., et al., Executive summary of the Stages of Reproductive Aging Workshop + 10: addressing the unfinished agenda of staging reproductive aging. J Clin Endocrinol Metab, 2012. 97(4): p. 1159-68.
  3. Burger, H.G., et al., A review of hormonal changes during the menopausal transition: focus on findings from the Melbourne Women's Midlife Health Project. Hum Reprod Update, 2007. 13(6): p. 559-65.
  4. Santoro, N., et al., Impaired folliculogenesis and ovulation in older reproductive aged women. J Clin Endocrinol Metab, 2003. 88(11): p. 5502-9.
  5. Hale, G.E., et al., Atypical estradiol secretion and ovulation patterns caused by luteal out-of-phase (LOOP) events underlying irregular ovulatory menstrual cycles in the menopausal transition. Menopause, 2009. 16(1): p. 50-9.
  6. Santoro, N., et al., Factors related to declining luteal function in women during the menopausal transition. J Clin Endocrinol Metab, 2008. 93(5): p. 1711-21.
  7. Santoro, N., et al., Menstrual Cycle Hormone Changes in Women Traversing Menopause: Study of Women's Health Across the Nation. J Clin Endocrinol Metab, 2017. 102(7): p. 2218-2229.
  8. Finkelstein, J.S., et al., Bone mineral density changes during the menopause transition in a multiethnic cohort of women. J Clin Endocrinol Metab, 2008. 93(3): p. 861-8.
  9. Paramsothy, P., et al., Duration of the menopausal transition is longer in women with young age at onset: the multiethnic Study of Women's Health Across the Nation. Menopause, 2017. 24(2): p. 142-149.
  10. Gold, E.B., et al., Factors related to age at natural menopause: longitudinal analyses from SWAN. Am J Epidemiol, 2013. 178(1): p. 70-83.
  11. Torgerson, D.J., R.E. Thomas, and D.M. Reid, Mothers and daughters menopausal ages: is there a link? Eur J Obstet Gynecol Reprod Biol, 1997. 74(1): p. 63-6.
  12. He, C. and J.M. Murabito, Genome-wide association studies of age at menarche and age at natural menopause. Mol Cell Endocrinol, 2014. 382(1): p. 767-79.
  13. Luborsky, J.L., et al., Premature menopause in a multi-ethnic population study of the menopause transition. Hum Reprod, 2003. 18(1): p. 199-206.
  14. Minino, A.M., et al., Deaths: final data for 2008. Natl Vital Stat Rep, 2011. 59(10): p. 1-126.
  15. Kannel, W.B., et al., Menopause and risk of cardiovascular disease: the Framingham study. Ann Intern Med, 1976. 85(4): p. 447-52.
  16. Chae, C.U. and C.A. Derby, The menopausal transition and cardiovascular risk. Obstet Gynecol Clin North Am, 2011. 38(3): p. 477-88.
  17. Hildreth, K.L., et al., Vascular dysfunction across the stages of the menopausal transition is associated with menopausal symptoms and quality of life. Menopause, 2018.
  18. El Khoudary, S.R., et al., Endogenous sex hormones impact the progression of subclinical atherosclerosis in women during the menopausal transition. Atherosclerosis, 2012. 225(1): p. 180-6.
  19. Moreau, K.L., et al., Endothelial function is impaired across the stages of the menopause transition in healthy women. J Clin Endocrinol Metab, 2012. 97(12): p. 4692-700.
  20. Hulley, S., et al., Randomized trial of estrogen plus progestin for secondary prevention of coronary heart disease in postmenopausal women. Heart and Estrogen/progestin Replacement Study (HERS) Research Group. JAMA, 1998. 280(7): p. 605-13.
  21. Effects of hormone replacement therapy on endometrial histology in postmenopausal women. The Postmenopausal Estrogen/Progestin Interventions (PEPI) Trial. The Writing Group for the PEPI Trial. JAMA, 1996. 275(5): p. 370-5.
  22. Anderson, G.L., et al., Effects of conjugated equine estrogen in postmenopausal women with hysterectomy: the Women's Health Initiative randomized controlled trial. JAMA, 2004. 291(14): p. 1701-12.
  23. Rossouw, J.E., et al., Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women's Health Initiative randomized controlled trial. Jama, 2002. 288(3): p. 321-33.
  24. Manson, J.E., et al., Menopausal Hormone Therapy and Long-term All-Cause and Cause-Specific Mortality: The Women's Health Initiative Randomized Trials. JAMA, 2017. 318(10): p. 927-938.
  25. Harman, S.M., et al., Arterial imaging outcomes and cardiovascular risk factors in recently menopausal women: a randomized trial. Ann Intern Med, 2014. 161(4): p. 249-60.
  26. Hodis, H.N., et al., Vascular Effects of Early versus Late Postmenopausal Treatment with Estradiol. N Engl J Med, 2016. 374(13): p. 1221-31.
  27. Woodard, G.A., et al., Lipids, menopause, and early atherosclerosis in Study of Women's Health Across the Nation Heart women. Menopause, 2011. 18(4): p. 376-84.
  28. Matthews, K.A., et al., Are changes in cardiovascular disease risk factors in midlife women due to chronological aging or to the menopausal transition? J Am Coll Cardiol, 2009. 54(25): p. 2366-73.
  29. El Khoudary, S.R., et al., Progression rates of carotid intima-media thickness and adventitial diameter during the menopausal transition. Menopause, 2013. 20(1): p. 8-14.
  30. Stefanick, M.L., et al., Effects of diet and exercise in men and postmenopausal women with low levels of HDL cholesterol and high levels of LDL cholesterol. N Engl J Med, 1998. 339(1): p. 12-20.
  31. Manson, J.E., et al., Estrogen plus progestin and the risk of coronary heart disease. N Engl J Med, 2003. 349(6): p. 523-34.
  32. Manson, J.E., et al., Estrogen therapy and coronary-artery calcification. N Engl J Med, 2007. 356(25): p. 2591-602.
  33. Wu, O., Postmenopausal hormone replacement therapy and venous thromboembolism. Gend Med, 2005. 2 Suppl A: p. S18-27.
  34. Canonico, M., et al., Hormone therapy and venous thromboembolism among postmenopausal women: impact of the route of estrogen administration and progestogens: the ESTHER study. Circulation, 2007. 115(7): p. 840-5.
  35. Canonico, M., et al., Postmenopausal Hormone Therapy and Risk of Stroke: Impact of the Route of Estrogen Administration and Type of Progestogen. Stroke, 2016. 47(7): p. 1734-41.
  36. Bergendal, A., et al., Risk of venous thromboembolism associated with local and systemic use of hormone therapy in peri- and postmenopausal women and in relation to type and route of administration. Menopause, 2016. 23(6): p. 593-9.
  37. Simon, J.A., et al., Venous thromboembolism and cardiovascular disease complications in menopausal women using transdermal versus oral estrogen therapy. Menopause, 2016. 23(6): p. 600-10.
  38. Adomaityte, J., M. Farooq, and R. Qayyum, Effect of raloxifene therapy on venous thromboembolism in postmenopausal women. A meta-analysis. Thromb Haemost, 2008. 99(2): p. 338-42.
  39. Hernandez, R.K., et al., Tamoxifen treatment and risk of deep venous thrombosis and pulmonary embolism: a Danish population-based cohort study. Cancer, 2009. 115(19): p. 4442-9.
  40. Christiansen, C., et al., Safety of bazedoxifene in a randomized, double-blind, placebo- and active-controlled Phase 3 study of postmenopausal women with osteoporosis. BMC Musculoskelet Disord, 2010. 11: p. 130.
  41. Cummings, S.R., et al., Epidemiology of osteoporosis and osteoporotic fractures. Epidemiol Rev, 1985. 7: p. 178-208.
  42. Riggs, B.L., et al., Rates of bone loss in the appendicular and axial skeletons of women. Evidence of substantial vertebral bone loss before menopause. J Clin Invest, 1986. 77(5): p. 1487-91.
  43. Shoback, D., et al., eds. Mineral metabolism and bone disease. Basic and Clinical Endocrinology, ed. F.S. Greenspan and D.G. Gardner. 2001, Lange Medical Books/McGraw Hill: New York. 237-333.
  44. Blake, G.M. and I. Fogelman, How important are BMD accuracy errors for the clinical interpretation of DXA scans? J Bone Miner Res, 2008. 23(4): p. 457-62.
  45. Force, U.S.P.S.T., et al., Screening for Osteoporosis to Prevent Fractures: US Preventive Services Task Force Recommendation Statement. JAMA, 2018. 319(24): p. 2521-2531.
  46. Camacho, P.M., et al., American Association of Clinical Endocrinologists and American College of Endocrinology Clinical Practice Guidelines for the Diagnosis and Treatment of Postmenopausal Osteoporosis - 2016. Endocr Pract, 2016. 22(Suppl 4): p. 1-42.
  47. Qaseem, A., et al., Treatment of Low Bone Density or Osteoporosis to Prevent Fractures in Men and Women: A Clinical Practice Guideline Update From the American College of Physicians. Ann Intern Med, 2017. 166(11): p. 818-839.
  48. Kanis, J.A., et al., A meta-analysis of the efficacy of raloxifene on all clinical and vertebral fractures and its dependency on FRAX. Bone, 2010. 47(4): p. 729-35.
  49. Puntila, E., et al., Leisure-time physical activity and rate of bone loss among peri- and postmenopausal women: a longitudinal study. Bone, 2001. 29(5): p. 442-6.
  50. Ross, A.C., et al., The 2011 report on dietary reference intakes for calcium and vitamin D from the Institute of Medicine: what clinicians need to know. J Clin Endocrinol Metab, 2011. 96(1): p. 53-8.
  51. Jackson, R.D., et al., Calcium plus vitamin D supplementation and the risk of fractures. N Engl J Med, 2006. 354(7): p. 669-83.
  52. Bostick, R.M., et al., Relation of calcium, vitamin D, and dairy food intake to ischemic heart disease mortality among postmenopausal women. Am J Epidemiol, 1999. 149(2): p. 151-61.
  53. Michaelsson, K., et al., Long term calcium intake and rates of all cause and cardiovascular mortality: community based prospective longitudinal cohort study. BMJ, 2013. 346: p. f228.
  54. Chung, M., et al., Vitamin D and calcium: a systematic review of health outcomes. Evid Rep Technol Assess (Full Rep), 2009(183): p. 1-420.
  55. Bolland, M.J., et al., Calcium and vitamin D supplements and health outcomes: a reanalysis of the Women's Health Initiative (WHI) limited-access data set. Am J Clin Nutr, 2011. 94(4): p. 1144-9.
  56. Bolland, M.J., et al., Concordance of Results from Randomized and Observational Analyses within the Same Study: A Re-Analysis of the Women's Health Initiative Limited-Access Dataset. PLoS One, 2015. 10(10): p. e0139975.
  57. Prentice, R.L., et al., Health risks and benefits from calcium and vitamin D supplementation: Women's Health Initiative clinical trial and cohort study. Osteoporosis International, 2013. 24: p. 567-80.
  58. Xiao, Q., et al., Dietary and supplemental calcium intake and cardiovascular disease mortality: the National Institutes of Health-AARP diet and health study. JAMA Intern Med, 2013. 173(8): p. 639-46.
  59. Paik, J.M., et al., Calcium supplement intake and risk of cardiovascular disease in women. Osteoporos Int, 2014. 25(8): p. 2047-56.
  60. Donneyong, M.M., et al., Risk of heart failure among postmenopausal women: a secondary analysis of the randomized trial of vitamin D plus calcium of the women's health initiative. Circ Heart Fail, 2015. 8(1): p. 49-56.
  61. Khaw, K.T., et al., Effect of monthly high-dose vitamin D supplementation on falls and non-vertebral fractures: secondary and post-hoc outcomes from the randomised, double-blind, placebo-controlled ViDA trial. Lancet Diabetes Endocrinol, 2017. 5(6): p. 438-447.
  62. Scragg, R., et al., Effect of Monthly High-Dose Vitamin D Supplementation on Cardiovascular Disease in the Vitamin D Assessment Study : A Randomized Clinical Trial. JAMA Cardiol, 2017. 2(6): p. 608-616.
  63. Eriksen, E.F., Treatment of osteopenia. Rev Endocr Metab Disord, 2012. 13(3): p. 209-23.
  64. Ettinger, B., et al., Reduction of vertebral fracture risk in postmenopausal women with osteoporosis treated with raloxifene: results from a 3-year randomized clinical trial. Multiple Outcomes of Raloxifene Evaluation (MORE) Investigators. JAMA, 1999. 282(7): p. 637-45.
  65. Cummings, S.R., et al., The effect of raloxifene on risk of breast cancer in postmenopausal women: results from the MORE randomized trial. Multiple Outcomes of Raloxifene Evaluation. JAMA, 1999. 281(23): p. 2189-97.
  66. Smith, C.L., et al., Breast-related effects of selective estrogen receptor modulators and tissue-selective estrogen complexes. Breast Cancer Res, 2014. 16(3): p. 212.
  67. Cummings, S.R., et al., Denosumab for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med, 2009. 361(8): p. 756-65.
  68. Finkelstein, J.S., et al., The effects of parathyroid hormone, alendronate, or both in men with osteoporosis. N Engl J Med, 2003. 349(13): p. 1216-26.
  69. Henriksen, K., et al., A randomized, double-blind, multicenter, placebo-controlled study to evaluate the efficacy and safety of oral salmon calcitonin in the treatment of osteoporosis in postmenopausal women taking calcium and vitamin D. Bone, 2016. 91: p. 122-9.
  70. Cummings, S.R., et al., Monitoring osteoporosis therapy with bone densitometry: misleading changes and regression to the mean. Fracture Intervention Trial Research Group. JAMA, 2000. 283(10): p. 1318-21.
  71. Col, N.F., et al., Duration of vasomotor symptoms in middle-aged women: a longitudinal study. Menopause, 2009. 16(3): p. 453-7.
  72. Politi, M.C., M.D. Schleinitz, and N.F. Col, Revisiting the duration of vasomotor symptoms of menopause: a meta-analysis. J Gen Intern Med, 2008. 23(9): p. 1507-13.
  73. Avis, N.E., et al., Duration of menopausal vasomotor symptoms over the menopause transition. JAMA Intern Med, 2015. 175(4): p. 531-9.
  74. Freeman, E.W., M.D. Sammel, and R.J. Sanders, Risk of long-term hot flashes after natural menopause: evidence from the Penn Ovarian Aging Study cohort. Menopause, 2014. 21(9): p. 924-32.
  75. Freedman, R.R., Menopausal hot flashes: mechanisms, endocrinology, treatment. J Steroid Biochem Mol Biol, 2014. 142: p. 115-20.
  76. Mittelman-Smith, M.A., et al., Role for kisspeptin/neurokinin B/dynorphin (KNDy) neurons in cutaneous vasodilatation and the estrogen modulation of body temperature. Proc Natl Acad Sci U S A, 2012. 109(48): p. 19846-51.
  77. Depypere, H., et al., Clinical evaluation of the NK3 receptor antagonist fezolinetant (a.k.a. ESN364) for the treatment of menopausal hot flashes. Maturitas, 2017. 103: p. 89-90.
  78. Bromberger, J.T., The menopausal transition increases the risk of depressive symptoms and depression diagnosis in women without a history of depression. Evid Based Ment Health, 2006. 9(4): p. 110.
  79. Cohen, L.S., et al., Risk for new onset of depression during the menopausal transition: the Harvard study of moods and cycles. Arch Gen Psychiatry, 2006. 63(4): p. 385-90.
  80. Freeman, E.W., et al., Associations of hormones and menopausal status with depressed mood in women with no history of depression. Arch Gen Psychiatry, 2006. 63(4): p. 375-82.
  81. Bromberger, J.T., et al., Major depression during and after the menopausal transition: Study of Women's Health Across the Nation (SWAN). Psychol Med, 2011. 41(9): p. 1879-88.
  82. Kravitz, H.M. and H. Joffe, Sleep during the perimenopause: a SWAN story. Obstet Gynecol Clin North Am, 2011. 38(3): p. 567-86.
  83. Bromberger, J.T., et al., Does risk for anxiety increase during the menopausal transition? Study of women's health across the nation. Menopause, 2013. 20(5): p. 488-95.
  84. Joffe, H., et al., Lifetime history of depression and anxiety disorders as a predictor of quality of life in midlife women in the absence of current illness episodes. Arch Gen Psychiatry, 2012. 69(5): p. 484-92.
  85. Kimura, D., Estrogen replacement therapy may protect against intellectual decline in postmenopausal women. Horm Behav, 1995. 29(3): p. 312-21.
  86. Shumaker, S.A., et al., Conjugated equine estrogens and incidence of probable dementia and mild cognitive impairment in postmenopausal women: Women's Health Initiative Memory Study. JAMA, 2004. 291(24): p. 2947-58.
  87. Greendale, G.A., et al., Effects of the menopause transition and hormone use on cognitive performance in midlife women. Neurology, 2009. 72(21): p. 1850-7.
  88. Gleason, C.E., et al., Effects of Hormone Therapy on Cognition and Mood in Recently Postmenopausal Women: Findings from the Randomized, Controlled KEEPS-Cognitive and Affective Study. PLoS Med, 2015. 12(6): p. e1001833; discussion e1001833.
  89. Manly, J.J., et al., Endogenous estrogen levels and Alzheimer's disease among postmenopausal women. Neurology, 2000. 54(4): p. 833-7.
  90. Tang, M.X., et al., Relative risk of Alzheimer disease and age-at-onset distributions, based on APOE genotypes among elderly African Americans, Caucasians, and Hispanics in New York City. Am J Hum Genet, 1996. 58(3): p. 574-84.
  91. Kawas, C., et al., A prospective study of estrogen replacement therapy and the risk of developing Alzheimer's disease: the Baltimore Longitudinal Study of Aging. Neurology, 1997. 48(6): p. 1517-21.
  92. Henderson, V.W., et al., Estrogen replacement therapy in older women. Comparisons between Alzheimer's disease cases and nondemented control subjects. Arch Neurol, 1994. 51(9): p. 896-900.
  93. Asthana, S., et al., High-dose estradiol improves cognition for women with AD: results of a randomized study. Neurology, 2001. 57(4): p. 605-12.
  94. Henderson, V.W., et al., Estrogen for Alzheimer's disease in women: randomized, double-blind, placebo-controlled trial. Neurology, 2000. 54(2): p. 295-301.
  95. Mulnard, R.A., et al., Estrogen replacement therapy for treatment of mild to moderate Alzheimer disease: a randomized controlled trial. Alzheimer's Disease Cooperative Study. JAMA, 2000. 283(8): p. 1007-15.
  96. McEwen, B.S., et al., Ovarian steroids and the brain: implications for cognition and aging. Neurology, 1997. 48(5 Suppl 7): p. S8-15.
  97. Gandy, S. and K. Duff, Post-menopausal estrogen deprivation and Alzheimer's disease. Exp Gerontol, 2000. 35(4): p. 503-11.
  98. Leiblum, S.R., et al., Hypoactive sexual desire disorder in postmenopausal women: US results from the Women's International Study of Health and Sexuality (WISHeS). Menopause, 2006. 13(1): p. 46-56.
  99. Johannes, C.B., et al., Distressing sexual problems in United States women revisited: prevalence after accounting for depression. J Clin Psychiatry, 2009. 70(12): p. 1698-706.
  100. Wierman, M.E., et al., Androgen therapy in women: a reappraisal: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab, 2014. 99(10): p. 3489-510.
  101. Davis, S.R., et al., Testosterone for low libido in postmenopausal women not taking estrogen. N Engl J Med, 2008. 359(19): p. 2005-17.
  102. Somboonporn, W., et al., Testosterone effects on the breast: implications for testosterone therapy for women. Endocr Rev, 2004. 25(3): p. 374-88.
  103. Hankinson, S.E. and A.H. Eliassen, Endogenous estrogen, testosterone and progesterone levels in relation to breast cancer risk. J Steroid Biochem Mol Biol, 2007. 106(1-5): p. 24-30.
  104. Simon, J.A., et al., Efficacy and safety of flibanserin in postmenopausal women with hypoactive sexual desire disorder: results of the SNOWDROP trial. Menopause, 2014. 21(6): p. 633-40.
  105. Fisher, W.A. and R.E. Pyke, Flibanserin Efficacy and Safety in Premenopausal Women With Generalized Acquired Hypoactive Sexual Desire Disorder. Sex Med Rev, 2017. 5(4): p. 445-460.
  106. Sakorafas, G.H., E. Krespis, and G. Pavlakis, Risk estimation for breast cancer development; a clinical perspective. Surg Oncol, 2002. 10(4): p. 183-92.
  107. Chlebowski, R.T., et al., Influence of estrogen plus progestin on breast cancer and mammography in healthy postmenopausal women: the Women's Health Initiative Randomized Trial. JAMA, 2003. 289(24): p. 3243-53.
  108. Stefanick, M.L., et al., Effects of conjugated equine estrogens on breast cancer and mammography screening in postmenopausal women with hysterectomy. JAMA, 2006. 295(14): p. 1647-57.
  109. Greendale, G.A., et al., Effects of estrogen and estrogen-progestin on mammographic parenchymal density. Postmenopausal Estrogen/Progestin Interventions (PEPI) Investigators. Ann Intern Med, 1999. 130(4 Pt 1): p. 262-9.
  110. Huo, C.W., et al., Mammographic density-a review on the current understanding of its association with breast cancer. Breast Cancer Res Treat, 2014. 144(3): p. 479-502.
  111. Bonnier, P., et al., Clinical and biologic prognostic factors in breast cancer diagnosed during postmenopausal hormone replacement therapy. Obstet Gynecol, 1995. 85(1): p. 11-7.
  112. Delgado, R.C. and D.M. Lubian Lopez, Prognosis of breast cancers detected in women receiving hormone replacement therapy. Maturitas, 2001. 38(2): p. 147-56.
  113. Anderson, G.L., et al., Conjugated equine oestrogen and breast cancer incidence and mortality in postmenopausal women with hysterectomy: extended follow-up of the Women's Health Initiative randomised placebo-controlled trial. Lancet Oncol, 2012. 13(5): p. 476-86.
  114. Committee on Practice, B.-G., Practice Bulletin Number 179: Breast Cancer Risk Assessment and Screening in Average-Risk Women. Obstet Gynecol, 2017. 130(1): p. e1-e16.
  115. Oeffinger, K.C., et al., Breast Cancer Screening for Women at Average Risk: 2015 Guideline Update From the American Cancer Society. JAMA, 2015. 314(15): p. 1599-614.
  116. Woolf, S.H., The 2009 breast cancer screening recommendations of the US Preventive Services Task Force. JAMA, 2010. 303(2): p. 162-3.
  117. Spiegelman, D., et al., Validation of the Gail et al. model for predicting individual breast cancer risk. J Natl Cancer Inst, 1994. 86(8): p. 600-7.
  118. Pruthi, S., R.E. Heisey, and T.B. Bevers, Chemoprevention for Breast Cancer. Ann Surg Oncol, 2015. 22(10): p. 3230-5.
  119. Fisher, B., et al., Tamoxifen for prevention of breast cancer: report of the National Surgical Adjuvant Breast and Bowel Project P-1 Study. J Natl Cancer Inst, 1998. 90(18): p. 1371-88.
  120. Vogel, V.G., et al., Effects of tamoxifen vs raloxifene on the risk of developing invasive breast cancer and other disease outcomes: the NSABP Study of Tamoxifen and Raloxifene (STAR) P-2 trial. JAMA, 2006. 295(23): p. 2727-41.
  121. Vogel, V.G., et al., Update of the National Surgical Adjuvant Breast and Bowel Project Study of Tamoxifen and Raloxifene (STAR) P-2 Trial: Preventing breast cancer. Cancer Prev Res (Phila), 2010. 3(6): p. 696-706.
  122. Kelly, C.M., et al., Selective serotonin reuptake inhibitors and breast cancer mortality in women receiving tamoxifen: a population based cohort study. BMJ, 2010. 340: p. c693.
  123. Goss, P.E., et al., Exemestane for breast-cancer prevention in postmenopausal women. N Engl J Med, 2011. 364(25): p. 2381-91.
  124. Cheung, A.M., et al., Bone density and structure in healthy postmenopausal women treated with exemestane for the primary prevention of breast cancer: a nested substudy of the MAP.3 randomised controlled trial. Lancet Oncol, 2012. 13(3): p. 275-84.
  125. Cuzick, J., et al., Anastrozole for prevention of breast cancer in high-risk postmenopausal women (IBIS-II): an international, double-blind, randomised placebo-controlled trial. Lancet, 2014. 383(9922): p. 1041-8.
  126. Sestak, I., et al., Changes in bone mineral density at 3 years in postmenopausal women receiving anastrozole and risedronate in the IBIS-II bone substudy: an international, double-blind, randomised, placebo-controlled trial. Lancet Oncol, 2014. 15(13): p. 1460-8.
  127. American College of, O. and P. Gynecologists Committee on Gynecologic, ACOG Committee Opinion No. 483: Primary and preventive care: periodic assessments. Obstet Gynecol, 2011. 117(4): p. 1008-15.
  128. Helfand, M. and C.C. Redfern, Clinical guideline, part 2. Screening for thyroid disease: an update. American College of Physicians. Ann Intern Med, 1998. 129(2): p. 144-58.
  129. Ladenson, P.W., et al., American Thyroid Association guidelines for detection of thyroid dysfunction. Arch Intern Med, 2000. 160(11): p. 1573-5.
  130. Portman, D.J., M.L. Gass, and P. Vulvovaginal Atrophy Terminology Consensus Conference, Genitourinary syndrome of menopause: new terminology for vulvovaginal atrophy from the International Society for the Study of Women's Sexual Health and the North American Menopause Society. Menopause, 2014. 21(10): p. 1063-8.
  131. Leiblum, S., et al., Vaginal atrophy in the postmenopausal woman. The importance of sexual activity and hormones. JAMA, 1983. 249(16): p. 2195-8.
  132. Rioux, J.E., et al., 17beta-estradiol vaginal tablet versus conjugated equine estrogen vaginal cream to relieve menopausal atrophic vaginitis. Menopause, 2000. 7(3): p. 156-61.
  133. Hendrix, S.L., et al., Effects of estrogen with and without progestin on urinary incontinence. JAMA, 2005. 293(8): p. 935-48.
  134. Henriksson, L., et al., A one-year multicenter study of efficacy and safety of a continuous, low-dose, estradiol-releasing vaginal ring (Estring) in postmenopausal women with symptoms and signs of urogenital aging. Am J Obstet Gynecol, 1996. 174(1 Pt 1): p. 85-92.
  135. Constantine, G., et al., Female sexual function improved with ospemifene in postmenopausal women with vulvar and vaginal atrophy: results of a randomized, placebo-controlled trial. Climacteric, 2015. 18(2): p. 226-32.
  136. Constantine, G.D., S.R. Goldstein, and D.F. Archer, Endometrial safety of ospemifene: results of the phase 2/3 clinical development program. Menopause, 2015. 22(1): p. 36-43.
  137. Kangas, L. and M. Unkila, Tissue selectivity of ospemifene: pharmacologic profile and clinical implications. Steroids, 2013. 78(12-13): p. 1273-80.
  138. Archer, D.F., et al., Treatment of pain at sexual activity (dyspareunia) with intravaginal dehydroepiandrosterone (prasterone). Menopause, 2015. 22(9): p. 950-63.
  139. Labrie, F., et al., Marked decline in serum concentrations of adrenal C19 sex steroid precursors and conjugated androgen metabolites during aging. J Clin Endocrinol Metab, 1997. 82(8): p. 2396-402.
  140. Treatment of menopause-associated vasomotor symptoms: position statement of The North American Menopause Society. Menopause, 2004. 11(1): p. 11-33.
  141. ACOG Practice Bulletin No. 141: management of menopausal symptoms. Obstet Gynecol, 2014. 123(1): p. 202-16.
  142. Weber, L. and H.L. Thacker, Paroxetine: a first for selective serotonin reuptake inhibitors - a new use: approved for vasomotor symptoms in postmenopausal women. Womens Health (Lond Engl), 2014. 10(2): p. 147-54.
  143. Loprinzi, C.L., et al., Venlafaxine in management of hot flashes in survivors of breast cancer: a randomised controlled trial. Lancet, 2000. 356(9247): p. 2059-63.
  144. Butt, D.A., et al., Gabapentin for the treatment of menopausal hot flashes: a randomized controlled trial. Menopause, 2008. 15(2): p. 310-8.
  145. Toulis, K.A., et al., Gabapentin for the treatment of hot flashes in women with natural or tamoxifen-induced menopause: a systematic review and meta-analysis. Clin Ther, 2009. 31(2): p. 221-35.
  146. Crandall, C.J., et al., Association of genetic variation in the tachykinin receptor 3 locus with hot flashes and night sweats in the Women's Health Initiative Study. Menopause, 2017. 24(3): p. 252-261.
  147. Prague, J.K., et al., Neurokinin 3 receptor antagonism as a novel treatment for menopausal hot flushes: a phase 2, randomised, double-blind, placebo-controlled trial. Lancet, 2017. 389(10081): p. 1809-1820.
  148. McCurry, S.M., et al., Telephone-Based Cognitive Behavioral Therapy for Insomnia in Perimenopausal and Postmenopausal Women With Vasomotor Symptoms: A MsFLASH Randomized Clinical Trial. JAMA Intern Med, 2016. 176(7): p. 913-20.
  149. National Institutes of, H., National Institutes of Health State-of-the-Science Conference statement: management of menopause-related symptoms. Ann Intern Med, 2005. 142(12 Pt 1): p. 1003-13.
  150. Santen, R.J., et al., Postmenopausal hormone therapy: an Endocrine Society scientific statement. J Clin Endocrinol Metab, 2010. 95(7 Suppl 1): p. s1-s66.
  151. Ettinger, B., et al., Cyclic hormone replacement therapy using quarterly progestin. Obstet Gynecol, 1994. 83(5 Pt 1): p. 693-700.

 

The Normal Menstrual Cycle and the Control of Ovulation

ABSTRACT

 

Menstruation is the cyclic, orderly sloughing of the uterine lining, in response to the interactions of hormones produced by the hypothalamus, pituitary, and ovaries. The menstrual cycle may be divided into two phases: (1) follicular or proliferative phase, and (2) the luteal or secretory phase. The length of a menstrual cycle is the number of days between the first day of menstrual bleeding of one cycle to the onset of menses of the next cycle. The median duration of a menstrual cycle is 28 days with most cycle lengths between 25 to 30 days (1-3. Patients who experience menstrual cycles that occur at intervals less than 21 days are termed polymenorrheic, while patients who experience prolonged menstrual cycles greater than 35 days, are termed oligomenorrheic. The typical volume of blood lost during menstruation is approximately 30 mL (4). Any amount greater than 80 mL is considered abnormal (4). The menstrual cycle is typically most irregular around the extremes of reproductive life (menarche and menopause) due to anovulation and inadequate follicular development (5-7). The luteal phase of the cycle is relatively constant in all women, with a duration of 14 days. The variability of cycle length is usually derived from varying lengths of the follicular phase of the cycle, which can range from 10 to 16 days. For complete coverage of this and related topics, please visit www.endotext.org.

 

THE FOLLICULAR PHASE (Fig.1)

Figure 1. Hormonal, Ovarian, endometrial, and basal body temperature changes and relations throughout the normal menstrual cycle. (From Carr BR, Wilson JD. Disorders of the ovary and female reproductive tract. In: Braunwald E, Isselbacher KJ, Petersdorf RG, et al, eds. Harrison's Principles of Internal Medicine. 11th ed. New York: McGraw-Hill, 1987: 1818-1837.

 

The follicular phase begins from the first day of menses until ovulation. Lower temperatures on a basal body temperature chart, and more importantly, the development of ovarian follicles, characterize this phase. Folliculogenesis begins during the last few days of the preceding menstrual cycle until the release of the mature follicle at ovulation.

 

Declining steroid production by the corpus luteum and the dramatic fall of inhibin A allows for follicle stimulating hormone (FSH) to rise during the last few days of the menstrual cycle (Fig. 2) (8). Another influential factor on the FSH level in the late luteal phase is related to an increase in GnRH pulsatile secretion secondary to a decline in both estradiol and progesterone levels (9). This elevation in FSH allows for the recruitment of a cohort of ovarian follicles in each ovary, one of which is destined to ovulate during the next menstrual cycle. Once menses ensues, FSH levels begin to decline due to the negative feedback of estrogen and the negative effects of inhibin B produced by the developing follicle (Fig. 2) (8, 10-12). FSH activates the aromatase enzyme in granulosa cells, which converts androgens to estrogen. A decline in FSH levels leads to the production of a more androgenic microenvironment within adjacent follicles to the growing dominant follicle. Also, the granulosa cells of the growing follicle secrete a variety of peptides that may play an autocrine/paracrine role in the inhibition of development of the adjacent follicles.

Figure 2. Inhibin level changes throughout the menstrual cycle. Inhibin B dominates the follicular phase of the cycle, while Inhibin A dominates the luteal phase.

 

Development of the dominant follicle has been described in three stages: (1) Recruitment, (2) Selection, and (3) Dominance (Fig.3). The recruitment stage takes place during days 1 through 4 of the menstrual cycle. During this stage, FSH leads to recruitment of a cohort of follicles from the pool of non-proliferating follicles. Between cycle days 5 and 7, selection of a follicle takes place whereby only one follicle is selected from the cohort of recruited follicles to ovulate, and the remaining follicles will undergo atresia. Anti-Müllerian hormone (AMH), a product of granulosa cells, is believed to play a role in the selection of the dominant follicle (13, 14). By cycle day 8, one follicle exerts its dominance by promoting its own growth and suppressing the maturation of the other ovarian follicles thus becoming the dominant follicle.

Figure 3. Time course for recruitment, selection, and ovulation of the dominant ovarian follicle (DF) with onset of atresia among other follicles of the cohort (N-1). (From Hodgen GD. The dominant ovarian follicle. Fertil Steril 1982; 38:281-300).

 

During the follicular phase, serum estradiol levels rise in parallel to the growth of follicle size as well as to the increasing number of granulosa cells. FSH receptors exist exclusively on the granulosa cell membranes. Increasing FSH levels during the late luteal phase leads to an increase in the number of FSH receptors and ultimately to an increase in estradiol secretion by granulosa cells. It is important to note that the increase in FSH receptor numbers is due to an increase in the population of granulosa cells and not due to an increase in the concentration of FSH receptors per granulosa cell. Each granulosa cell has approximately 1500 FSH receptors by the secondary stage of follicular development and FSH receptor numbers remains relatively constant for the remainder of development (15). The rise in estradiol secretion appears to increase the total number of estradiol receptors on the granulosa cells (16). In the presence of estradiol, FSH stimulates the formation of LH receptors on granulosa cells allowing for the secretion of small quantities of progesterone and 17-hydroxyprogesterone (17-OHP) which may exert a positive feedback on the estrogen- primed pituitary to augment luteinizing hormone (LH) release (17). FSH also stimulates several steroidogenic enzymes including aromatase, and 3β-hydroxysteroid dehydrogenase (3β-HSD) (18, 19). In table 1, the production rates of sex steroids during the follicular phase, luteal phase, and at the time of ovulation are presented.

 

 

Table 1. Production Rate of Sex Steroids in Women at Different Stages of the Menstrual Cycle
DAILY PRODUCTION RATE
SEX STEROIDS* Early
Follicular 
Preovulatory  Mid-luteal
Progesterone (mg) 1 4 25
17α-Hydroxyprogesterone (mg) 0.5 4 4
Dehydroepiandrosterone (mg) 7 7 7
Androstenedione (mg) 2.6 4.7 3.4
Testosterone (µg) 144 171 126
Estrone (µg) 50 350 250
Estradiol (µg) 36 380 250
From Baird DT. Fraser IS. Blood production and ovarian secretion rates of esuadiol-17β and estrone in women throughout the menstrual cycle. J Clin Endocrinol Metab 38: l009-1017. 1974. @ The Endocrine Society.
*Values are expressed in milligrams or micrograms per 24 hours.

 

In contrast to granulosa cells, LH receptors are located on theca cells during all stages of the menstrual cycle. LH principally stimulates androstenedione production, and to a lesser degree testosterone production in theca cells. In the human, androstenedione is then transported to the granulosa cells where it is aromatized to estrone and finally converted to estradiol by 17-β-hydroxysteroid dehydrogenase type I. This is known as the two-cell, two-gonadotropin hypothesis of regulation of estrogen synthesis in the human ovary (Fig. 4).

Figure 4. Two-cell, two-gonadotropin hypothesis of regulation of estrogen synthesis in the human ovary. Adapted by Carr, BR. Diseases of the ovary and Reproductive Tract. In Wilson JD, Foster DW, Kronenberg HM, Larsen PR, eds. Williams Textbook of Endocrinology 9th edition. WB Saunders, Philadelphia, p.751-817.

 

In the ovary, the primordial follicles are surrounded by a single layer of granulosa cells and are arrested in the diplotene stage of the first meiotic division. After puberty, each primordial follicle enlarges and develops into a preantral follicle. The preantral follicle is now surrounded by several layers of granulosa cells as well as by theca cells. The preantral follicle is the first stage of FSH receptivity, as now the follicle has acquired FSH receptors. The preantral follicle then develops a cavity and is now known as an antral follicle. Finally, it becomes a preovulatory follicle on its way towards ovulation. Due to the presence of 5α-reductase, preantral and early antral follicles produce more androstenedione and testosterone in relation to estrogens (20). 5α-reductase is the enzyme responsible for converting testosterone to dihydrotestosterone (DHT). Once testosterone has been 5α-reduced, DHT cannot be aromatized. However, the dominant follicle is able to secrete large quantities of estrogen, primarily estradiol, due to high levels of CYP19 (aromatase). This shift from an androgenic to an estrogenic follicular microenvironment may play an important role in selection of the dominant follicle from those follicles that will become atretic.

 

As mentioned earlier, development of the follicle to the preantral stage is gonadotropin independent, and any follicular growth beyond this point will require gonadotropin interaction. Gonadotropin secretion is regulated by gonadotropin releasing hormone (GnRH), steroid hormones, and various peptides released by the dominant follicle. Also, as mentioned earlier, FSH is elevated during the early follicular phase and then begins to decline until ovulation. In contrast, LH is low during the early follicular phase and begins to rise by the mid-follicular phase due to the positive feedback from the rising estrogen levels. For the positive feedback effect of LH release to occur, estradiol levels must be greater than 200 pg/mL for approximately 50 hours in duration (21). Gonadotropins are normally secreted in a pulsatile fashion from the anterior pituitary, and the frequency and amplitude of the pulses vary according to the phase of the menstrual cycle (Table 2). During the early follicular phase, LH secretion occurs at a pulse frequency of 60 to 90 minutes with relatively constant pulse amplitude. During the late follicular phase prior to ovulation, the pulse frequency increases and the amplitude may begin to increase. In most women, the LH pulse amplitude begins to increase after ovulation takes place (22).

 

 

Table 2. Mean (SEM) Luteinizing Hormone Secretory Burst Characteristics During Phases of the Menstrual Cycle*
NUMBER
(24 hr)
PERODICITY (min) AMPLITUDE** (mlU/ml/min) HALF-DURATIONS (min) LH HALF-LIFE (min) TOTAL DAILY SECRETION (mlU/ml/24 hr)
Early follicular 175±1.4a 80 ± 3a 0.43 ± 0.02a 6.5 ± 1.0a 131 ± 13a 49 ± 6a
Late follicular 26.9±1.6b  53 ± 1b 0.70 ± 0.03b 3.5 ± 0.9b 128 ± 12a 56 ± 8a
Midluteal 10.1±1.0c  177 ± 15# 0.26 ± 0.02c# 11.0 ± 1.1e 103 ± 7a 52 ± 4a
395 ± 37d# 0.95 ± 0.05d#
*Entries in each column identified by a, b, c, d differ significantly (Duncan's multiple-range test, P <.05). Periodicity is intersecretory burst interval. LH, Luteinizing hormone.
**Duration of the deconvolution-resolved LH secretory burst at half-maximal amplitude.
#Maximal rate of LH secretion attained with the deconvolution-resolved LH secretory burst. The midluteal phase has been divided into small (less than 0.65 mIU/ml/min) and large (greater than 0.65 mIU/ml/min) secretory burst amplitudes.
Data from Sollenberger MJ, Carlsen EC, Johnson ML, et al. Specific physiological regulation of LH secretory events throughout the human menstrual cycle. New insights into the pulsatile mode of gonadotropin release. J Neuroendocrinol 2:845, 1990.

 

There are numerous substances found in follicular fluid, such as steroids, pituitary hormones, plasma proteins, proteoglycans and non-steroidal ovarian factors, which regulate the microenvironment of the ovary and regulate steroidogenesis in granulosa cells. Growth factors such as insulin-like growth factor 1 and 2 (IGF1, IGF2) and epidermal growth factor (EGF) are recognized as playing important roles in oocyte development and maturation (23-25). The concentration of ovarian steroids is much higher in follicular fluid in comparison to plasma concentrations. There are 2 populations of antral follicles: (1) large follicles, which are greater than 8mm in diameter, and (2) small follicles, which are less than 8mm. In the large follicles, the concentrations of FSH, estrogen, and progesterone are high while prolactin concentration is low. In the small follicles, prolactin and androgen levels are higher compared to large antral follicles (26).

 

OVULATION

 

Ovulation occurs approximately 10-12 hours after the LH peak (Fig. 5) (27). The LH surge is initiated by a dramatic rise of estradiol produced by the preovulatory follicle (Fig. 6). To produce the critical concentration of estradiol needed to initiate the positive feedback, the dominant follicle is almost always >15mm in diameter on ultrasound (28). The beginning of the LH surge occurs roughly 34 to 36 hours prior to ovulation and is a relatively precise predictor for timing ovulation (Fig. 5) (29). The LH surge stimulates luteinization of the granulosa cells and stimulates the synthesis of progesterone responsible for the midcycle FSH surge. Also, the LH surge stimulates resumption of meiosis and the completion of reduction division in the oocyte with the release of the first polar body. It has been demonstrated in cultured granulosa cells that spontaneous luteinization can occur in the absence of LH. It is hypothesized that the inhibitory effects of factors such as oocyte maturation inhibitor or luteinization inhibitor are overcome at ovulation (30).

Figure 5. The onset of LH surge usually precedes ovulation by 36 hours. The peak, on the other hand preceded ovulation by 10-12 hours.

 

Figure 6. Changes in gonadotropins and ovarian steroids at midcycle, just prior to ovulation. The initiation of LH surge is at time 0. Abbreviations: E2, estrogen; P, progesterone (From Hoff JD, Quigley ME, Yen SCC. Hormonal dynamics at midcycle: A re-evaluation. J Clin Endocrinol Metab. 57:792, 1983.

 

Prostaglandins and proteolytic enzymes, such as collagenase and plasmin, are increased in response to LH and progesterone. Although the precise mechanism is not known, proteolytic enzymes and prostaglandins are activated and digest collagen in the follicular wall, leading to an explosive release of the oocyte-cumulus complex (31). Prostaglandins may also stimulate ovum release by stimulation of smooth muscle within the ovary. The point of the dominant follicle closest to the ovarian surface where this digestion occurs is called the stigma. There is no evidence to support the theory that follicular rupture occurs as a result of increased follicular pressure, although precise measurements precisely at rupture have not been performed (32). In a recent report, laparoscopic visualization of human ovulation during an operative procedure was documented. The authors report visualizing a follicular area called the stigma which was protruding like a bleb from the surface, containing viscous yellow fluid evaginating into the peritoneal cavity (33). In humans, ovulation probably occurs randomly from either ovary during any given cycle. Of interest, some studies have suggested that ovulation occurs more commonly from the right ovary and right sided ovulation carries a higher potential for pregnancy (34). The concentrations of prostaglandins E and F series and hydroxyeicosatetraenoic acid (HETE) reach a peak level in follicular fluid just prior to ovulation (35, 36). Prostaglandins may stimulate proteolytic enzymes while HETEs may stimulate angiogenesis and hyperemia (37). Patients treated with high dose prostaglandin synthetase inhibitors such as Indocin, can have a block in prostaglandin production and effectively block follicular rupture (38-40). This gives rise to what is known as the luteinized, unruptured follicle syndrome and it presents in fertile and infertile patients equally (41). Therefore, infertility patients are advised to avoid taking prostaglandin synthetase inhibitors, as well as cyclo-oxygenase (COX) inhibitors, especially around the time of ovulation (40). A schematic diagram illustrating the proposed mechanisms involved in follicular rupture is presented in Figure 7.

Figure 7. Proposed mechanisms involved in follicular rupture. From Tsafriri A, Chun S-Y. Ovulation. In: Adashi E, Rock JA, Rosenwaks Z. Reproductive Endocrinology, Surgery and Technology. Philadelphia: Lippincott-Raven, 1996:236-249.

 

Estradiol levels fall dramatically immediately prior to the LH peak. This may be due to LH downregulation of its own receptor or because of direct inhibition of estradiol synthesis by progesterone. Progesterone is also responsible for stimulating the midcycle rise in FSH. Elevated FSH levels at this time are thought to free the oocyte from follicular attachments, stimulate plasminogen activator, and increase granulosa cell LH receptors. The mechanism causing the postovulatory fall in LH is unknown. The decline in LH may be due to the loss of the positive feedback effect of estrogen, due to the increasing inhibitory feedback effect of progesterone, or due to a depletion of LH content of the pituitary from downregulation of GnRH receptors (42).

 

LUTEAL PHASE

 

This phase is usually 14 days long in most women. After ovulation, the remaining granulosa cells that are not released with the oocyte continue to enlarge, become vacuolated in appearance, and begin to accumulate a yellow pigment called lutein. The luteinized granulosa cells combine with the newly formed theca-lutein cells and surrounding stroma in the ovary to become what is known as the corpus luteum. The corpus luteum is a transient endocrine organ that predominantly secretes progesterone, and its primary function is to prepare the estrogen primed endometrium for implantation of the fertilized ovum. The basal lamina dissolves and capillaries invade into the granulosa layer of cells in response to secretion of angiogenic factors by the granulosa and thecal cells (43). Eight or nine days after ovulation, approximately around the time of expected implantation, peak vascularization is achieved. Figure 8 demonstrates a corpus luteum as seen on transvaginal ultrasound. Note the increased blood flow seen surrounding the corpus luteum as seen with Doppler evaluation. This time also corresponds to peak serum levels of progesterone and estradiol. The central cavity of the corpus luteum may also accumulate with blood and become a hemorrhagic corpus luteum. The life span of the corpus luteum depends upon continued LH support. Corpus luteum function declines by the end of the luteal phase unless human chorionic gonadotropin is produced by a pregnancy. If pregnancy does not occur, the corpus luteum undergoes luteolysis under the influence of estradiol and prostaglandins and forms a scar tissue called the corpus albicans.

Figure 8. Corpus luteum as seen on transvaginal ultrasound. On the right image, note the Doppler flow indicating vascular flow surrounding the structure.

 

Estrogen levels rise and fall twice during the menstrual cycle. Estrogen levels rise during the mid-follicular phase and then drop precipitously after ovulation. This is followed by a secondary rise in estrogen levels during the mid-luteal phase with a decrease at the end of the menstrual cycle. The secondary rise in estradiol parallels the rise of serum progesterone and 17α-hydroxyprogesterone levels. Ovarian vein studies confirm that the corpus luteum is the site of steroid production during the luteal phase (44).

 

The mechanism by which the corpus luteum regulates steroid secretion is not completely understood. Regulation may be determined in part by LH secretory pattern and LH receptors or variations in the levels of the enzymes regulating steroid hormone production, such as 3β-HSD, CYP17, CYP19, or side chain cleavage enzyme. The number of granulosa cells formed during the follicular phase and the amount of readily available LDL cholesterol may also play a role in steroid regulation by the corpus luteum. The luteal cell population consists of at least two cell types, the large and the small cells (45). Small cells are thought to have been derived from thecal cells while the large cells from granulosa cells. The large cells are more active in steroidogenesis and are influenced by various autocrine/paracrine factors such as inhibin, relaxin, and oxytocin (46, 47).

 

In studies looking into the mechanisms regulating the menstrual cycle, LH was established as the primary luteotropic agent in a cohort of hypophysectomized women (48). After induction of ovulation, the amount of progesterone secreted and the length of the luteal phase is dependent on repeated LH injections. Administration of LH or HCG during the luteal phase can extend corpus luteum function for an additional two weeks (49).

 

The secretion of progesterone and estradiol during the luteal phase is episodic, and correlates closely with pulses of LH secretion (Fig. 9) (50). The frequency and amplitude of LH secretion during the follicular phase regulates subsequent luteal phase function and is consistent with the regulatory role of LH during the luteal phase (51). Reduced levels of FSH during the follicular phase can lead to a shortened luteal phase and the development of a smaller corpus lutea (52). Also, the life span of the corpus luteum can be reduced by continuous LH administration during the follicular or luteal phase, reduced LH concentration, decreased LH pulse frequency, or decreased LH pulse amplitude (53-55). The role of other luteotropic factors such as prolactin, oxytocin, inhibin and relaxin is still unclear (56, 57).

Figure 9. Episodic secretion of LH (top) and progesterone (bottom) during the luteal phase of a woman. Abbreviations: LH, luteinizing hormone: P, progesterone E2, estradiol; LH + 8, LH surge plus 8 days. (From Filicori M, Butler JP, Crowley WF Jr. Neuroendocrine regulation of the corpus luteum in the human. J Clin Invest. 73:1638 1984.

 

The corpus luteum function begins to decline 9-11 days after ovulation. The exact mechanism of how the corpus luteum undergoes its demise is unknown. Estrogen is believed to play a role in the luteolysis of the corpus luteum (58). Estradiol injected into the ovary bearing the corpus luteum induces luteolysis while no effect is noted after estradiol injection of the contralateral ovary (56). However, the absence of estrogen receptors in human luteal cells does not support the role of endogenous estrogen in corpus luteum regression (59). Prostaglandin F2α appears to be luteolytic in nonhuman primates and in studies of women (60, 61). Prostaglandin F2α exerts its effects via the synthesis of endothelin-1, which inhibits steroidogenesis and stimulates the release of a growth factor, tumor necrosis factor alpha (TNFα), which induces cell apoptosis (62). Oxytocin and vasopressin exert their luteotropic effects via an autocrine/paracrine mechanism (63). Luteinizing hormone's ability to downregulate its own receptor may also play a role in termination of the luteal phase. Finally, Matrix metalloproteinases also appear to play a role in luteolysis (64).

 

Not all hormones undergo marked fluctuations during the normal menstrual cycle. Androgens, glucocorticoids, and pituitary hormones, excluding LH and FSH, undergo only minimal fluctuation (65-68). Due to extra-adrenal 21-hyroxylation of progesterone, plasma levels of deoxycorticosterone are increased during the luteal phase (69, 70).

 

HORMONAL EFFECTS ON THE REPRODUCTIVE TRACT

 

Endometrium

 

The effects of varying concentrations of estrogen and progesterone throughout the course of the menstrual cycle have characteristic effects on the endometrium (Fig. 10) (71). The endometrial changes that occur can be visualized with sonography (Fig. 11). The characteristic endometrial changes also allow for histologic dating.  Histologic dating is most accurately accomplished by performing an endometrial biopsy 2-3 days prior to expected menstruation. The proliferative phase is more difficult to date accurately in comparison to the luteal phase. The glands during the proliferative phase are narrow, tubular, and some mitosis and pseudostratification is present. The endometrium thickness is usually between 0.5 and 5mm. In a classical 28-day menstrual cycle, ovulation occurs on day 14. On cycle day 16, the glands take on a more pseudostratified appearance with glycogen accumulating at the basal portion of the glandular epithelium and some nuclei are displaced to the midportion of the cells. In a formalin fixed specimen, glycogen is solubilized resulting in the characteristic basal vacuolization at the base of the endometrial cells. This finding confirms the formation of a functional, progesterone producing, corpus luteum. In the luteal phase, progesterone decreases the biologic activity of estradiol on the endometrium by: (1) decreasing the concentration of estradiol receptors, (2) increasing the enzymatic activity of 17β-hydroxysteroid dehydrogenase type II, the enzyme responsible for the conversion of estradiol to estrone, and (3) by increasing the activity of estrone sulfotransferase (72, 73).

Figure 10. Dating of the Endometrium. From Noyes RW, Hertig AW, Rock J. Dating the endometrial biopsy. Fertil Steril 1950; 1:3.

Figure 11. Characteristic sonographic endometrial changes seen throughout the menstrual cycle.

 

On cycle day 17, the endometrial glands become more tortuous and dilated. On cycle day 18, the vacuoles in the epithelium decrease in size and are frequently located next to the nuclei. Also, glycogen is now found at the apex of the endometrial cells. By cycle day 19, the pseudostratification and vacuolation almost completely disappear and intraluminal secretions become present. On cycle day 21 or 22, the endometrial stroma begins to become edematous. On cycle day 23, stromal cells surrounding the spiral arterioles begin to enlarge and stromal mitoses become apparent. On cycle day 24, predecidual cells appear around the spiral arterioles and stromal mitoses become more apparent. On cycle day 25, the predecidua begins to differentiate under the surface epithelium. On cycle day 27, there is a marked lymphocytic infiltration and the upper endometrial stroma appears as a solid sheet of well-developed decidua-like cells. On cycle day 28, menstruation begins.

 

In 2004, Chan et al., were the first to confirm that stem cells were present in human endometrium (73A). Subsequent research has involved characterization of the different types of endometrial stem cells (73B). Importantly, menstrual fluid may be an easily accessible source for certain types of endometrial stem cells (73C). This may lead to advancements in the treatment of many gynecologic disorders including endometriosis and Asherman syndrome as well as non-gynecologic disorders such as neurologic and cardiac disorders (73B).

 

Cervix

 

The mucous secreting glands of the endocervix are affected by the changes in steroid hormone concentration. Immediately after menstruation, the cervical mucous is scant and viscous. During the late follicular phase, under the influence of rising estradiol levels, the cervical mucous becomes clear, copious and elastic. The quantity of cervical mucous increases 30 fold compared to the early follicular phase (74). The stretchability or elasticity of the cervical mucous can be evaluated between two glass slides and recorded as the spinnbarkeit. If examined under the microscope, the cervical mucous will display a characteristic ferning or palm-leaf arborization appearance. After ovulation, as progesterone levels rise, the cervical mucous once again becomes thick, viscous and opaque and the quantity produced by the endocervical cells decreases.

 

Vagina

 

The changes in hormonal levels of estrogen and progesterone also have characteristic effects on the vaginal epithelium. During the early follicular phase, exfoliated vaginal epithelial cells have vesicular nuclei and are basophilic. During the late follicular phase, and the influence of the rising estradiol level, the vaginal epithelial cells display pyknotic nuclei and are acidophilic (75). As progesterone rises during the luteal phase, the acidophilic cells decrease in number and are replaced by an increasing number of leukocytes.

 

MENSTRUATION

 

In the absence of a pregnancy, steroid hormone levels begin to fall due to declining corpus luteum function. Progesterone withdrawal results in increased coiling and constriction of the spiral arterioles. This eventually results in tissue ischemia due to decreased blood flow to the superficial endometrial layers, the spongiosa and compacta. The endometrium releases prostaglandins that cause contractions of the uterine smooth muscle and sloughing of the degraded endometrial tissue. The release of prostaglandins may be due to decreased stability of lysosomal membranes in the endometrial cells (76). Infusions of prostaglandin F2α in women during the luteal phase has been shown to induce endometrial necrosis and bleeding (77). The use of prostaglandin synthetase inhibitors decreases the amount of menstrual bleeding and can be used as therapy in women with excessive menstrual bleeding or menorrhagia. Menstrual fluid is composed of desquamated endometrial tissue, red blood cells, inflammatory exudates, and proteolytic enzymes. Within two days after the start of menstruation and while endometrial shedding is still occurring, estrogen produced by the growing follicles starts to stimulate the regeneration of the surface endometrial epithelium. The estrogen secreted by the growing ovarian follicles, causes prolonged vasoconstriction enabling the formation of a clot over the denuded endometrial vessels (78). Also, the regeneration and remodeling of the uterine connective tissue is regulated in part by the matrix metalloproteinase (MMP) system (79).

 

The average duration of menstrual flow is between four to six days, but the normal range in women can be from as little as two days up to eight days. As mentioned earlier, the average amount of menstrual blood loss is 30 mL and greater than 80 mL is considered abnormal [4].

 

MENSTRUAL DISORDERS

 

Apart from conditions of abnormal menstruation, certain disorders are increased in women when compared to men. These conditions are thought to be related to hormone differences as well as hormone changes throughout the menstrual cycle. Increased autoimmune conditions, such as rheumatoid arthritis or systemic lupus erythematosus, are believed to be related to estrogen enhancement of humoral immunity (80). Other researchers also describe higher vulnerability for drug abuse during phases of the menstrual cycle when estradiol levels are high (81).

 

SUMMARY

 

The length of a menstrual cycle is the number of days between the first day of menstrual bleeding of one cycle to the onset of menses of the next cycle. The median duration of a menstrual cycle is 28 days with most cycle lengths between 25 to 30 days. The menstrual cycle may be divided into two phases: (1) follicular or proliferative phase, and (2) the luteal or secretory phase. The follicular phase begins from the first day of menses until ovulation. The development of ovarian follicles characterizes this phase. The LH surge is initiated by a dramatic rise of estradiol produced by the preovulatory follicle and results in subsequent ovulation. The LH surge stimulates luteinization of the granulosa cells and stimulates the synthesis of progesterone responsible for the midcycle FSH surge. Also, the LH surge stimulates resumption of meiosis and the completion of reduction division in the oocyte with the release of the first polar body. The luteal phase is 14 days long in most women. If the corpus luteum is not rescued by pregnancy, it will undergo atresia. The resultant progesterone withdrawal results in menses. The average duration of menstrual flow is between four and six days, but the normal range in women can be from as little as two days up to eight days. The average amount of menstrual blood   is 30ml, and over 60 ml is considered abnormal.

 

REFERENCES

 

  1. Treloar, A.E., et al., Variation of the human menstrual cycle through reproductive life. Int J Fertil, 1967. 12(1 Pt 2): p. 77-126.
  2. Vollman, R.F., The Menstrual Cycle. 1977, WB Saunders: Philadelphia.
  3. Presser, H.B., Temporal data relating to the human menstrual cycle, in Biorhythms and Human Reproduction, M. Ferin, et al., Editors. 1974, John Wiley and Sons: New York. p. 145-160.
  4. Hallberg, L., et al., Menstrual blood loss--a population study. Variation at different ages and attempts to define normality. Acta Obstet Gynecol Scand, 1966. 45(3): p. 320-351.
  5. Apter, D., et al., Follicular growth in relation to serum hormonal patterns in adolescent compared with adult menstrual cycles. Fertil Steril, 1987. 47(1): p. 82-88.
  6. Fraser, I.S., et al., Pituitary gonadotropins and ovarian function in adolescent dysfunctional uterine bleeding. J Clin Endocrinol Metab, 1973. 37(3): p. 407-414.
  7. Lenton, E.A., et al., Normal variation in the length of the follicular phase of the menstrual cycle: effect of chronological age. Br J Obstet Gynaecol, 1984. 91(7): p. 681-684.
  8. Groome, N.P., et al., Measurement of dimeric inhibin B throughout the human menstrual cycle. J Clin Endocrinol Metab, 1996. 81(4): p. 1401-1405.
  9. Welt, C.K., et al., Control of follicle-stimulating hormone by estradiol and the inhibins: critical role of estradiol at the hypothalamus during the luteal-follicular transition. J Clin Endocrinol Metab, 2003. 88(4): p. 1766-1771.
  10. Tsafriri, A., Local nonsteroidal regulators of ovarian function, in The physiology of reproduction, E. Knobil, J.D. Neill, and et al., Editors. 1994, Raven: New York. p. 817-860.
  11. Sawetawan, C., et al., Inhibin and activin differentially regulate androgen production and 17 alpha-hydroxylase expression in human ovarian thecal-like tumor cells. J Endocrinol, 1996. 148(2): p. 213-221.
  12. Welt, C.K., et al., Frequency modulation of follicle-stimulating hormone (FSH) during the luteal-follicular transition: evidence for FSH control of inhibin B in normal women. J Clin Endocrinol Metab, 1997. 82(8): p. 2645-2652.
  13. Durlinger, A.L., et al., Control of primordial follicle recruitment by anti-Mullerian hormone in the mouse ovary. Endocrinology, 1999. 140(12): p. 5789-96.
  14. Hampl, R., M. Snajderova, and T. Mardesic, Antimullerian hormone (AMH) not only a marker for prediction of ovarian reserve. Physiol Res, 2011. 60(2): p. 217-23.
  15. Amsterdam, A. and S. Rotmensch, Structure-function relationships during granulosa cell differentiation. Endocr.Rev, 1987. 8(3): p. 309-337.
  16. Nimrod, A., G.F. Erickson, and K.J. Ryan, A specific FSH receptor in rat granulosa cells: properties of binding in vitro. Endocrinology, 1976. 98(1): p. 56-64.
  17. Fink, G., Gonadotropin secretion and its control, in The physiology of reproduction, E. Knobil, J.D. Neill, and et al., Editors. 1988, Raven: New York. p. 1349-1377.
  18. Zeleznik, A.J., A.R. Midgley, Jr., and L.E. Reichert, Jr., Granulosa cell maturation in the rat: increased binding of human chorionic gonadotropin following treatment with follicle-stimulating hormone in vivo. Endocrinology, 1974. 95(3): p. 818-825.
  19. Erickson, G.F., C. Wang, and A.J. Hsueh, FSH induction of functional LH receptors in granulosa cells cultured in a chemically defined medium. Nature, 1979. 279(5711): p. 336-338.
  20. McNatty, K.P., et al., Metabolism of androstenedione by human ovarian tissues in vitro with particular reference to reductase and aromatase activity. Steroids, 1979. 34(4): p. 429-443.
  21. Young, J.R. and R.B. Jaffe, Strength-duration characteristics of estrogen effects on gonadotropin response to gonadotropin-releasing hormone in women. II. Effects of varying concentrations of estradiol. J Clin Endocrinol Metab, 1976. 42(3): p. 432-442.
  22. Reame, N., et al., Pulsatile gonadotropin secretion during the human menstrual cycle: evidence for altered frequency of gonadotropin-releasing hormone secretion. J Clin Endocrinol Metab, 1984. 59(2): p. 328-337.
  23. Zhou, J. and C. Bondy, Anatomy of the human ovarian insulin-like growth factor system. Biol Reprod, 1993. 48(3): p. 467-482.
  24. Maruo, T., et al., Expression of epidermal growth factor and its receptor in the human ovary during follicular growth and regression. Endocrinology, 1993. 132(2): p. 924-931.
  25. Thierry van Dessel, H.J., et al., Serum and follicular fluid levels of insulin-like growth factor I (IGF-I), IGF-II, and IGF-binding protein-1 and -3 during the normal menstrual cycle. J Clin Endocrinol Metab, 1996. 81(3): p. 1224-31.
  26. Hillier, S.G., et al., Intraovarian sex steroid hormone interactions and the regulation of follicular maturation: aromatization of androgens by human granulosa cells in vitro. J Clin Endocrinol Metab, 1980. 50(4): p. 640-647.
  27. Pauerstein, C.J., et al., Temporal relationships of estrogen, progesterone, and luteinizing hormone levels to ovulation in women and infrahuman primates. Am J Obstet Gynecol, 1978. 130(8): p. 876-886.
  28. Cahill, D.J., et al., Onset of the preovulatory luteinizing hormone surge: diurnal timing and critical follicular prerequisites. Fertil Steril, 1998. 70(1): p. 56-59.
  29. Hoff, J.D., M.E. Quigley, and S.S. Yen, Hormonal dynamics at midcycle: a reevaluation. J Clin Endocrinol Metab, 1983. 57(4): p. 792-796.
  30. Channing, C.P., et al., Ovarian follicular and luteal physiology, in International Review of Physiology, R.O. Greep, Editor. 1980, University Park Press: Baltimore. p. 117.
  31. Espey, L.L. and H. Lipner, Ovulation, in The Physiology of Reproduction, E. Knobil and J.D. Neill, Editors. 1994, Raven: New York. p. 725.
  32. Espey, L.L., Ovarian proteolytic enzymes and ovulation. Biol Reprod, 1974. 10(2): p. 216-235.
  33. Lousse, J.C. and J. Donnez, Laparoscopic observation of spontaneous human ovulation. Fertil Steril, 2008. 90(3): p. 833-834.
  34. Fukuda, M., et al., Right-sided ovulation favours pregnancy more than left-sided ovulation. Hum Reprod, 2000. 15(9): p. 1921-1926.
  35. Lumsden, M.A., et al., Changes in the concentration of prostaglandins in preovulatory human follicles after administration of hCG. J Reprod Fertil, 1986. 77(1): p. 119-124.
  36. Espey, L.L., et al., Ovarian hydroxyeicosatetraenoic acids compared with prostanoids and steroids during ovulation in rats. Am J Physiol., 1991. 260(2 Pt 1): p. E163-E169.
  37. Sherman, B.M., J.H. West, and S.G. Korenman, The menopausal transition: analysis of LH, FSH, estradiol, and progesterone concentrations during menstrual cycles of older women. J Clin Endocrinol Metab, 1976. 42(4): p. 629-636.
  38. O'Grady, J.P., et al., The effects of an inhibitor of prostaglandin synthesis (indomethacin) on ovulation, pregnancy, and pseudopregnancy in the rabbit. Prostaglandins, 1972. 1(2): p. 97-106.
  39. Killick, S. and M. Elstein, Pharmacologic production of luteinized unruptured follicles by prostaglandin synthetase inhibitors. Fertil Steril, 1987. 47(5): p. 773-777.
  40. Pall, M., B.E. Friden, and M. Brannstrom, Induction of delayed follicular rupture in the human by the selective COX-2 inhibitor rofecoxib: a randomized double-blind study. Hum Reprod, 2001. 16(7): p. 1323-1328.
  41. Doody, K.J. and B.R. Carr, Diagnosis and treatment of luteal dysfunction, in Ovarian Endocrinology, S.G. Hillier, Editor. 1991, Blackwell Scientific: London. p. 260.
  42. Katt, J.A., et al., The frequency of gonadotropin-releasing hormone stimulation determines the number of pituitary gonadotropin-releasing hormone receptors. Endocrinology, 1985. 116(5): p. 2113-2115.
  43. Koos, R.D., Potential relevance of angiogenic factors to ovarian physiology. Semin Reprod Endocrinol, 1989. 7 p. 29.
  44. Niswender, G.D. and T.M. Nett, The corpus luteum and its control in infraprimate species, in The Physiology of Reproduction, E. Knobil and J.D. Neill, Editors. 1994, Raven: New York. p. 781.
  45. Retamales, I., et al., Morpho-functional study of human luteal cell subpopulations. Hum Reprod, 1994. 9(4): p. 591-596.
  46. Khan-Dawood, F.S., et al., Human corpus luteum secretion of relaxin, oxytocin, and progesterone. J Clin Endocrinol Metab, 1989. 68(3): p. 627-631.
  47. Carr, B.R., et al., The effect of transforming growth factor-beta on steroidogenesis and expression of key steroidogenic enzymes with a human ovarian thecal-like tumor cell model. Am J Obstet Gynecol, 1996. 174(4): p. 1109-1116.
  48. Vande Wiele, R.L., et al., Mechanisms regulating the menstrual cycle in women. Recent.Prog.Horm.Res, 1970. 26: p. 63-103.
  49. Segaloff, A., W.H. Sternberg, and C.J. Gaskill, Effects of luteotropic dose of chorionic gonadotropin in women. J Clin Endocrinol Metab, 1951. 11(9): p. 936-944.
  50. Filicori, M., J.P. Butler, and W.F. Crowley, Jr., Neuroendocrine regulation of the corpus luteum in the human. Evidence for pulsatile progesterone secretion. J Clin Invest, 1984. 73(6): p. 1638-1647.
  51. McNeely, M.J. and M.R. Soules, The diagnosis of luteal phase deficiency: a critical review. Fertil Steril, 1988. 50(1): p. 1-15.
  52. Stouffer, R.L. and G.D. Hodgen, Induction of luteal phase defects in rhesus monkeys by follicular fluid administration at the onset of the menstrual cycle. J Clin Endocrinol Metab, 1980. 51(3): p. 669-671.
  53. Sheehan, K.L., R.F. Casper, and S.S. Yen, Luteal phase defects induced by an agonist of luteinizing hormone-releasing factor: a model for fertility control. Science, 1982. 215(4529): p. 170-172.
  54. Keyes, P.L. and M.C. Wiltbank, Endocrine regulation of the corpus luteum. Annu.Rev Physiol., 1988. 50: p. 465-482.
  55. Cooke, I.D., The corpus luteum. Hum Reprod, 1988. 3(2): p. 153-156.
  56. Auletta, F.J. and A.P. Flint, Mechanisms controlling corpus luteum function in sheep, cows, nonhuman primates, and women especially in relation to the time of luteolysis. Endocr.Rev, 1988. 9(1): p. 88-105.
  57. Hodgen, G.D., Surrogate embryo transfer combined with estrogen-progesterone therapy in monkeys. Implantation, gestation, and delivery without ovaries. JAMA, 1983. 250(16): p. 2167-2171.
  58. Gore, B.Z., B.V. Caldwell, and L. Speroff, Estrogen-induced human luteolysis. J Clin Endocrinol Metab, 1973. 36(3): p. 615-617.
  59. Iwai, T., et al., Immunohistochemical localization of oestrogen receptors and progesterone receptors in the human ovary throughout the menstrual cycle. Virchows Arch A Pathol Anat Histopathol, 1990. 417(5): p. 369-375.
  60. Wentz, A.C. and G.S. Jones, Transient luteolytic effect of prostaglandin F2alpha in the human. Obstet Gynecol, 1973. 42(2): p. 172-181.
  61. Schoonmaker, J.N., et al., Estradiol-induced luteal regression in the rhesus monkey: evidence for an extraovarian site of action. Endocrinology, 1982. 110(5): p. 1708-1715.
  62. Shikone, T., et al., Apoptosis of human corpora lutea during cyclic luteal regression and early pregnancy. J Clin Endocrinol Metab, 1996. 81(6): p. 2376-2380.
  63. Khan-Dawood, F.S., J.C. Huang, and M.Y. Dawood, Baboon corpus luteum oxytocin: an intragonadal peptide modulator of luteal function. Am J Obstet Gynecol, 1988. 158(4): p. 882-891.
  64. Young, K.A., J.D. Hennebold, and R.L. Stouffer, Dynamic expression of mRNAs and proteins for matrix metalloproteinases and their tissue inhibitors in the primate corpus luteum during the menstrual cycle. Mol Hum Reprod, 2002. 8(9): p. 833-840.
  65. Judd, H.L. and S.S. Yen, Serum androstenedione and testosterone levels during the menstrual cycle. J Clin Endocrinol Metab, 1973. 36(3): p. 475-481.
  66. Genazzani, A.R., et al., Pattern of plasma ACTH, hGH, and cortisol during menstrual cycle. J Clin Endocrinol Metab, 1975. 41(3): p. 431-437.
  67. Carr, B.R., et al., Plasma levels of adrenocorticotropin and cortisol in women receiving oral contraceptive steroid treatment. J Clin Endocrinol Metab, 1979. 49(3): p. 346-349.
  68. Carr, B.R. and J.D. Wilson, Disorders of the ovary and the female reproductive tract, in Harrison's Principles of Internal Medicine, E. Braunwald, et al., Editors. 1987, McGraw-Hill: New York. p. 1818.
  69. Casey, M.L. and P.C. MacDonald, Extraadrenal formation of a mineralocorticosteroid: deoxycorticosterone and deoxycorticosterone sulfate biosynthesis and metabolism. Endocr.Rev, 1982. 3(4): p. 396-403.
  70. Parker, C.R., Jr., et al., Plasma concentrations of 11-deoxycorticosterone in women during the menstrual cycle. Obstet Gynecol, 1981. 58(1): p. 26-30.
  71. Noyes, R.W., A.T. Hertig, and J. Rock, Dating the endometrial biopsy. Am J Obstet Gynecol, 1975. 122(2): p. 262-263.
  72. Lessey, B.A., et al., Immunohistochemical analysis of human uterine estrogen and progesterone receptors throughout the menstrual cycle. J Clin Endocrinol Metab, 1988. 67(2): p. 334-340.
  73. Tseng, L. and H.C. Liu, Stimulation of arylsulfotransferase activity by progestins in human endometrium in vitro. J Clin Endocrinol Metab, 1981. 53(2): p. 418-421.

73A.     Chan RWS, Schwab KE, Gargett CE: Clonogenicity of human endometrial epithelial and stromal cells. Biol Reprod 2004;70:1738–50.

73B.    Gargett CE, Schwab KE, Deane JA: Endometrial stem/progenitor cells: the first 10 years. Hum Reprod Update 2015;22:dmv051.

73C.    Patel AN, Park E, Kuzman M, Benetti F, Silva FJ, Allickson JG: Multipotent menstrual blood stromal stem cells: isolation, characterization, and differentiation. Cell Transplant 2008 [cited 2018 Mar 13];17:303–11

  1. Rebar, R.E., Practical evaluation of hormonal status, in Reproductive Endocrinology: Physiology, Pathophysiology and Clinical Management, S.S.C. Yen and R.B. Jaffe, Editors. 1991, WB Saunders: Philadelphia. p. 830.
  2. Gaudefroy, M., Cytologic criteria of estrogen effect. Acta Cytol, 1958(2): p. 347.
  3. Henzl, M.R., et al., Lysosomal concept of menstrual bleeding in humans. J Clin Endocrinol Metab, 1972. 34(5): p. 860-875.
  4. Turksoy, R.N. and H.S. Safaii, Immediate effect of prostaglandin F2alpha during the luteal phase of the menstrual cycle. Fertil Steril, 1975. 26(7): p. 634-637.
  5. Edman, C.D., The effects of steroids on the endometrium. Semin Reprod Endocrinol, 1983. 1(3): p. 179.
  6. Curry, T.E., Jr. and K.G. Osteen, Cyclic changes in the matrix metalloproteinase system in the ovary and uterus. Biol Reprod, 2001. 64(5): p. 1285-1296.
  7. Cutolo, M., Gender and the rheumatic diseases: Epidemiological evidence and possible biologic mechanisms. Annals of the Rheumatic Diseases, 2003. 62: p. 3-3.
  8. Anker, J.J. and M.E. Carroll, Females are more vulnerable to drug abuse than males: evidence from preclinical studies and the role of ovarian hormones. Curr Top Behav Neurosci, 2011. 8: p. 73-96.

 

 

 

 

Thyroxine Poisoning

CLINICAL RECOGNITION

 

A massive L-Thyroxine (T4) overdose may be accidentally and unintentionally ingested, most commonly by children and adolescents. It may occur intentionally in young and older adults in an attempt to lose weight, with suicidal intentions, or for undeclared purposes.  In some localities thyroxine may be obtained at drugstores without prescription (mostly in the generic form). In some reports thyroxine preparations by a pharmacist had an erroneous LT4 dosage. Thyroid hormone pills used to treat hypothyroid dogs typically contains a much higher dose of thyroid hormone and if mistakenly taken by humans can lead to thyroxine poisoning.

 

Ingested thyroxine, which is itself probably of modest physiologic significance, is rapidly partially converted to triiodothyronine (T3), the active form of thyroid hormone. Both thyroxine and triiodothyronine levels in serum rise within 1-2 hours of ingestion. Agents that inhibit T4>T3 conversion provide one approach to treatment. In children and adolescents, the clinical course is often very mild. Patients in this age range may ingest a full flask of LT4 with 90-100 tablets (100 or 150 mcg/tablet). Rarely, the overdose is discovered immediately and the patient is brought to the hospital 6-12 hours after the ingestion. At this time, the common clinical signs and symptoms include nervousness, insomnia, mild tremor of hands, tachycardia, mild elevation of body temperature, blood pressure elevation, and loose stools.  Rarely more serious late effects occur, including coma, convulsions, and acute psychosis. Cardiac effects aside from tachycardia are seldom seen in young adults but may occur in middle age and older adults, with reported arrhythmias and acute myocardial infarction. However, only one fatality has been reported. Interestingly the onset of symptoms and signs (Table 1) may be delayed for up to 3 to 10 days and does not correlate closely with plasma levels of serum total T4 and total T3. Medical consensus has indicated that serious symptoms are less frequent in children even though children usually have higher mean plasma levels of T4 and T3 than adults for the same overdose of LT4 ingested. One-time ingestion of up to 3 mg thyroxine rarely causes symptoms in adult or children. As already mentioned serious complications are not common, but they can appear days after ingestion, and therefore the patients should be closely monitored.

 

TABLE 1: SYMPTOMS AND SIGNS AFTER INGESTION OF LT4

Severe toxicity is quite rare in children.

Common-effects:

Nervousness

Insomnia

Mild elevation of temperature

Blood pressure elevation

Loose stools

Rare symptoms:

Comma

Convulsions

Acute psychosis

Thyroid storm

Tachycardia, arrhythmias

 

DIAGNOSIS and DIFFERENTIAL

 

Elevated levels of total and free T4 and T3 have been described with suppressed serum TSH levels and otherwise typically a normal biochemical profile (Table 2). The half-life of serum T4 may be shortened. In one study the half-life of LT4 was 5.7 days which is slightly shorter than the usual half-life of L-thyroxine. In one report total serum T3 levels reached the normal range five days after ingestion of 9.9 mg of LT4 (99 tablets of 100 mcg), although free T4 levels were still elevated. In many cases, there is a progressive rise in both serum total T4 and total T3 levels in the first 24 hours following the overdose, caused by continued absorption of the ingested LT4.

 

Table 2: Biochemical Changes After Ingestion of LT4

Elevated serum total T4 and T3

Suppressed serum TSH

Elevated Free T4 and Free T3

Normal biochemical profile

 

THERAPY

 

Therapeutic recommendations are made based only in the review of the available literature concerning a relatively large number of patients, most of them children. Acute levothyroxine overdose is much more common in children compared to adolescents and adults. Therapeutic options are related to the time elapsed after the ingestion of a large number of tablets of L-thyroxine and the actual beginning of emergency therapy (Table 3). Acute massive doses of L-thyroxine typically have a mild clinical course that can be controlled by activated charcoal, or possibly cholestyramine, propranolol, dexamethasone, and supporting measures, with close medical evaluation. Rarely critical cardiac conditions, coma, seizures will follow massive doses of L-Thyroxine.

 

If more than a few hours of ingestion of LT-4 tablets have elapsed, most probably the tablets have travelled from the gastric cavity to duodenum. Moreover, gastric lavage is difficult to conduct in small children. One way to confirm the presence of LT-4 tablets in the gastric cavity is endoscopy, easily conducted in many hospitals and emergency rooms. LT-4 tablets are dissolved by the gastric juice, but there are no data about the rate of dissolution of a large number of tablets of LT-4. Most probably LT-4 would not be entirely dissolved by the gastric juice and may not be absorbed in the duodenum (normally about 10-15%) but would be absorbed in the jejuno-ileum (normally about 53% of absorption of LT4).

 

Emetics both local (Ipecac) or central agents (apomorphine) should be avoided.

 

Administration of activated charcoal is a common practice in many drug overdoses and is an agent that can prevent absorption of several drugs from the gastro-intestinal system. However, in many reports repeated doses of activated charcoal were ineffective in accelerating the elimination of levothyroxine, probably due to high uptake in the duodenum and jejuno-ileum.

 

Hemoperfusion using activated charcoal is a rather complicated procedure but has been reported to be highly effective in decreasing total serum levels. It should be reserved for adult patients with severe intoxication by very large doses of thyroxine and the same applies to plasmapheresis which has been seldom used.  

 

Cholestyramine, an ion-exchange resin (Questran ®), can be administered in the usual dose of 4 grams every 8 hours orally. This drug binds thyroxine and enhances its elimination.

 

Glucocorticoids (Dexamethasone 4 mg orally) decrease the conversion of LT4 to T3, the active hormone. Sodium Ipodate (oral cholecystographic agent) has also been used for blocking the conversion of LT4 to T3, but it is no longer generally available.

 

Beta-blockers such as propranolol, are useful to ameliorate the metabolic effects of thyroid hormone, mostly on the cardiac system (controlling tachycardia, preventing arrhythmias). Seizures may be treated with phenytoin and phenobarbital. Propylthiouracil (PTU) might be used for blocking the conversion of T4 to T3 but may have very limited usefulness in the presence of a large load of LT4.  

Hemodialysis has been used in severe cases, but it is probably of limited value since both T3 and T4 are highly protein-bound.

 

TABLE 3: TREATMENT OF INGESTION OF A MASSIVE DOSE OF L-THYROXINE

Gastric lavage (within hours of ingestion).

Emetic agents (not advised)

Propranolol (10-40 mg 3 times daily)

Activated Charcoal (1g/kg p.o.)

Dexamethasone (4 mg p.o. daily)

Sodium ipodate, if available

Cholestyramine (4g every 8h p.o.)

Propylthiouracil (PTU) (May inhibit conversion of T4>T3)

Activated charcoal hemoperfusion

Plasmapheresis (seldom necessary)

Hemodialysis (probably of limited value)

Thyroid storm: demands treatment in an Intensive Care Unit.

 

FOLLOW-UP

 

Patients should be monitored for several days to be sure that serum T4 and T3 levels are falling

 

GUIDELINES

 

None applicable.

 

REFERENCES

 

Shilo L, Kovatz S, Hadari R, Weiss E, Nabriski D, Shenkman L. Massive thyroid hormone overdose: kinetics, clinical manifestations and management, Israel med Ass J 2002; 4:209-299. http://www.ncbi.nlm.nih.gov/pubmed/12001709

 

Kreisner E, Lutzky M, Gross JL. Charcoal hemoperfusion in the treatment of levothyroxine intoxication. Thyroid 2010, 20:209-212. http://www.ncbi.nlm.nih.gov/pubmed/20151829 De Luis

 

DA, Duenas A, Abad L, Aller R. Light symptoms following a high dose intentional L-Thyroxine ingestion treated with cholestyramine. Horm Res 2002; 67:61-62. http://www.ncbi.nlm.nih.gov/pubmed/12006723

 

De Groot LJ, Bartalena L. Thyroid Storm. In: De Groot LJ, Chrousos G, Dungan K, Feingold KR, Grossman A, Hershman JM, Koch C, Korbonits M, McLachlan R, New M, Purnell J, Rebar R, Singer F, Vinik A, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000-.

2015 Apr 12. PMID: 25905165

Cellular Action of Thyroid Hormone

ABSTRACT

Thyroid hormones (THs) regulate growth, development, metabolism. This chapter aims to provide a comprehensive overview of the molecular and cellular mechanism(s) for intracellular signaling by TH. At the cellular level, THs bind to thyroid hormone receptors (TRs) that are members of the nuclear hormone receptor family.  TRs act as ligand-activated transcription factors that bind to their cognate thyroid hormone response elements (TREs) in the promoters of target genes. TRs regulate gene transcription by employing TR-interacting protein complexes containing coactivators (CoAs) or  corepressors (CoRs). Coactivator and corepressor complexes include histone modifying enzymes known as histone acetyltransferases (HATs) or histone deacetylases  (HDACs), respectively, that induce epigenetic changes in the chromatin structure of target gene promoters to enhance or repress the transcriptional efficiency of RNA polymerase on TH-responsive genes. TRs also can mediate transcriptional effects indirectly by binding to other transcription factors or activating cell signaling cascades.  Additionally, emerging evidence suggests that THs may bind to cell membrane proteins other than TRs to activate cell signaling pathways. The important role of TH on metabolism, has  spawned interest the pharmacological use of TH and its analogs/metabolites for the treatment of metabolic diseases such as hypercholestronemia, hypertriglycerimia obesity, and non-alcoholic fatty liver disease (NAFLD).

 

INTRODUCTION

The thyroid hormones (THs, thyroxine (T4) and triiodothyronine (T3)) have important effects on development, growth, and metabolism (1-3). Some of the most prominent effects of TH occur during fetal development and early childhood. In humans, the early developmental role of TH is illustrated by the distinctive clinical features of cretinism observed in iodine-deficient areas. In childhood, lack of TH can cause delayed growth. However, in this latter case, many of the effects of TH may be metabolic rather than developmental, as growth is restored rapidly after the institution of TH treatment. In adults, the primary effects of THs are manifested by alterations in metabolism. These effects include changes in oxygen consumption, protein, carbohydrate, lipid, and vitamin metabolism. The clinical features of hypothyroidism and hyperthyroidism emphasize the pleiotropic effects of these hormones on many different pathways and target organs.

 

At the clinical level, identification of quantitative markers of TH action has been difficult (4). At the extreme ends of the clinical spectrum, which extends from hypothyroidism to hyperthyroidism, the diagnosis of a thyroid abnormality is usually apparent. Clinical suspicion of a thyroid abnormality can be confirmed using laboratory tests for THs and thyroid stimulating hormone (TSH). However, more subtle forms of thyroid dysfunction, such as subclinical hypothyroidism or hyperthyroidism, pose a greater challenge. Although the level of circulating TSH provides a sensitive and quantitative indicator of TH action at the level of the hypothalamic-pituitary axis, there are few reliable peripheral or intracellular markers of TH action (5,6). The effect of TH on basal metabolism has been re-evaluated using measurements of resting energy expenditure (REE). In hypothyroid patients taking varying levels of TH replacement, there is a strong inverse correlation between REE and the TSH level (6). Nevertheless, TSH remains the most sensitive and useful clinical indicator of TH action. As discussed below, tissue-selective metabolism of THs, and variable tissue sensitivity to their effects, underscores the need to develop additional markers of TH activity in peripheral tissues.

 

Since the initial description of TH effects on metabolic rate more than 100 years ago (7), many theories have been proposed to explain its mechanism of hormone action. The proposed models include: uncoupling oxidative phosphorylation, stimulation of energy expenditure by the activation of Na+-K+ ATPase activity, and direct modulation of TH transporters and enzymes in the plasma membrane and mitochondria (8). Recently, there has been increasing evidence for non-genomic actions (see later under non-genomic actions of TH) (8); however, the major effects of TH occur via nuclear receptors that mediate changes in gene expression.

 

In 1966, Tata proposed that TH increased gene expression with attendant increases in protein synthesis and enzyme activity (9). In 1972, high affinity nuclear binding sites for TH were documented (Kd approximately 10-10 M for T3) (10,11). The receptor-binding affinity of various THs and analogues correlated with their biologic potencies, consistent with the view that most biologic effects are mediated via the nuclear receptor (11– 14). Over the past 25 years, there has been a dramatic surge of new information on TH action resulting from the cloning of the TH receptors (15,16), the identification of regulatory DNA elements in TH responsive genes (1-3), the generation of TR isoform knockout mice (17,18), and the discovery and phenotype characterization of patients with mutations in TRα and TRβ (5,19,20). In this chapter, we will focus on our current understanding of nuclear TH receptor action.

BINDING OF THs TO NUCLEAR RECEPTORS

In many respects, T4can be regarded as a prohormone for the more potent hormone, T3. Most of the TH bound to receptors is in the form of T3, either secreted into the circulation by the thyroid gland or derived from T4to T3 conversion by 5' monodeiodinases (see Chapter 3C). There are three distinct deiodinases- type I, type II, and type III (21,22). The distribution and regulation of these enzymes can have important effects on TH action. For example, Type II deiodinase has high affinity for T4(Kd in the nanomolar range) and is found primarily in the pituitary gland, brain, and brown fat where conversion of T4to T3 modulates the intracellular concentration of T3. Thus, tissues that contain type II deiodinase can respond differently to a given circulating concentration of T4(by intracellular conversion to T3) than organs that only can respond to T3(23,24). Additionally, it appears that both type 1 and type II deiodinase regulate the circulating T4and T3levels (25). Recently, MCT8, OATP-1, and System L amino acid transporters have been identified as TH transporters which regulate T4and T3uptake into cells (26,27). Mutations in the former have been involved in a number of syndromes of x-linked mental retardation and neurologic deterioration (27,28).

 

T3binds to its receptors with approximately 10 fold higher affinity than T4. The dissociation constants for liver nuclear receptors measured in vitro are 2 x 10-9M for T4and 2 x 10-10M for T3(1,2). Nuclear receptors are approximately 75% saturated with TH in brain and pituitary and 50% saturated with TH in liver and kidney. It is notable that the extent of TH receptor occupancy varies in different tissues, providing a mechanism for alterations in circulating TH levels to alter receptor activity. In contrast to the related steroid hormone receptors, TRs are mostly nuclear both in the absence and presence of TH (1,2,30). In fact, TH receptors are tightly associated with chromatin (1-3,30), consistent with their proposed role as DNA-binding proteins that regulate gene expression.

CLONING, STRUCTURE, AND EXPRESSION OF TH RECEPTORS

Cloning of TRs

TH receptors (TRs) were first cloned in 1986 and belong to the nuclear hormone receptor superfamily that includes the glucocorticoid, estrogen, progesterone, androgen, aldosterone, vitamin D, retinoic acid (RARs), retinoid X (RXRs) and "orphan" (unknown ligand and/or DNA target) receptors (1,2,15,16,30-33). TRs are the cellular homologs of v-erbA, a viral oncogene product involved in chick erythroblastosis. TRs are encoded at two genomic loci (α and β) located on human chromosomes 17 and 3, respectively and their gene products result in two major isoforms, TRα and TRβ .

Structure of TRs

Like other members of the nuclear receptor superfamily, the TRs have a central DNA-binding domain (DBD) and a carboxy-terminal ligand-binding domain (LBD) (Figure 3d-1). The two major TR isoforms have high amino acid sequence homology in their respective DBDs and LBDs. Dimerization domains also are found in both DBDs and LBDs. The amino-terminal regions are more variable between TRα and TRβ (1-3), and contain ligand-independent activation domains. In contrast, multiple sub-regions are located in the LBD for ligand-dependent transcriptional activation and basal repression of target genes.

Figure 1. Functional domains of the TH receptor (TR). The TH receptor (TR) is depicted schematically. The zinc finger DNA-binding domain (DBD) is denoted along with the carboxy terminal ligand-binding domain (LBD). Other functional domains and interaction sites are indicated.

The DNA-binding domains of the nuclear receptors are comprised of two distinct zinc fingers that are separated by a 15-17 amino acid linker sequence. The crystal structure of the DNA-binding domains of the TH receptor and its heterodimeric partner, RXR has been determined. The two heterodimer partners interact with a direct repeat of the receptor binding site in a head-to-tail manner (34-37).

 

A small stretch of amino acids at the base of the first finger (referred to as the P-box) dictates the DNA sequence specificity of the receptor (35-39). The P-box sequence of the TH receptor is shared by other receptors that bind to similar or identical DNA recognition sites (AGGTCA). The underlined amino acids in the P-box (EGCKG) of the TH receptor are also found in the retinoic acid receptors, the retinoic acid X receptors, the rev-erbA protein, the vitamin D receptor, and NGFI-B. Of note, the steroid hormone receptors have a different P box sequence and bind as homodimers to a different consensus DNA half-site sequence (AGAACA). The region between the DBD and LBD is called the hinge region and contains the nuclear localization signal, typically a basic amino acid‑rich sequence, first described in viral nuclear proteins.

 

X‑ray crystallographic studies of the liganded rat TRα-1 show that TH is embedded in a hydrophobic "pocket" lined by discontinuous stretches of amino acid sequences within the LBD. Additionally, there are several hydrophobic interfaces within the LBD that contribute to the TR homo- and heterodimerization with RXR (39). There are twelve amphipathic helices in the LBD and specific helices among them provide the critical contact surfaces for protein-protein interactions with co-activators and co-repressors (helices 3,5,6,12 and 3,4,5,6, respectively) (40-43). Ligand-binding to TR causes a major conformational change in the LBD, particularly in helix 12. This, in turn, facilitates TR discrimination between co-activators and co-repressors (see below).

Splicing variants of TRs

The carboxy-terminal hormone-binding domain of the TRα gene is alternatively-spliced to generate several protein products (Figure 3d-2, below). One variant, referred to as α-2, is identical to TRα-1 through the first 370 amino acids, but then its sequence diverges completely, owing to splicing of alternate exons (44-47). Another splicing variant, referred to as TRvII or α-3, is similar to α-2 except that it lacks the first 39 amino acids found in the unique region of α-2 (45). α -2 cannot bind TH because of the replacement of critical amino acids at the extreme carboxy-terminal end of the protein due to alternative splicing (48), and thus cannot mediate ligand-dependent gene transcription (49– 51). The amino acid replacements in α-2 also alter its dimerization properties and reduce DNA-binding affinity (52-55). The α-2 splicing variant is highly expressed in many tissues such as brain, testis, kidney, and brown fat, but its function remains poorly understood (56). The α-2 isoform has been proposed to be an endogenous inhibitor of TH receptor function as it inhibits TRα and β activity in transient gene expression assays (44,54). The mechanism by which α-2 antagonizes TR action is controversial. Some studies indicate that α-2 competes for active receptor complexes at DNA target sites (57,58). Other studies indicate that α-2 inhibits TR activity independent of DNA-binding (59). It is likely that the inhibitory effects of α-2 involve more than one mechanism. Amino acid substitutions in the carboxy-terminal region of α-2 also prevent its interactions with transcriptional corepressors (see below) (55), and may provide an explanation as to why α-2 is not a more potent inhibitor of TR activity. Additionally, the phosphorylation state of α-2 may modulate its inhibitory activity (60). Given the foregoing features, the TRα-1 and α-2 system represents one of the few examples in mammals whereby multiple mRNAs generated by alternative splicing encode proteins that are antagonistic to each other.

Figure 2. TH receptor isoforms. The TH receptors (TR) β and α are expressed from separate genes. Each TR gene can be expressed as distinct isoforms, reflecting the use of alternate promoters and exons. The central zinc finger DNA-binding region is indicated and unique domains are shown by distinct patterns of shading. The TRβ-2 isoform, which is expressed predominantly in the pituitary and hypothalamus, contains a unique amino-terminus. The TRα-2 isoform contains unique carboxy-terminal sequences that eliminate hormone binding. The DNA- and T3-binding properties and transcriptional activity of the various isoforms are shown at the right.

A receptor-like molecule, Rev-erbA, is, surprisingly, encoded on the opposite strand of the TRα gene locus (61, 62). Rev-erbA mRNA contains a 269-nucleotide stretch which is complementary to the α-2 mRNA due to its transcription from the DNA strand opposite of that used to generate TRα-1 and α-2. This protein also is a member of the nuclear hormone receptor superfamily, and is highly expressed in adipocytes and muscle cells. Rev-erbA, contains a DBD that is homologous to the TR DBD. However, Rev-erbA does not bind TH and its putative LBD has minimal homology with other nuclear hormone receptors. Since no cognate ligand has been identified for Rev-erbA, it is categorized as an "orphan nuclear receptor. " It can act as a transcriptional repressor for nuclear hormone receptors and other transcription factors (63-65). Since Rev-erbA shares an exonic segment of the bidirectionally transcribed TRα gene, it is possible that it modulates the expression or splicing of TRα-1 and α-2 (66, 67) as parallel increases in rev-erbA mRNA and TRα-1 mRNA expression occur relative to α2 mRNA expression.

 

The major variant of the TRβ gene, TRβ-2, has a different amino-terminus than TRβ 1 (Figure 3d-2, above) (68). The distinct amino-terminal region of the TRβ-2 is due to transcription from a tissue-specific promoter. The function of the amino-terminus of the TH receptor is not known, but it likely plays a role in transcriptional control (69,70). The TRβ-1 and TRβ-2 isoforms function similarly in most transient gene expression assays (69, 71), although differences in the transcriptional activities of the TRβ 1 and TRβ 2 isoforms have been noted with respect to certain target genes (69-72). It is likely that tissue-restricted expression of the TRβ-2 isoform contributes to unique patterns of TR expression, which in turn, may modulate target gene regulation.

 

Recently, short isoforms of TRα and TRβ have been described (73, 74). The novel TRα isoforms arise from translational start sites in the 7th intron and yield shortened TRα 1 and α 2 isoforms that have dominant negative activity on WT TR. Novel short TRβ isoforms arise from alternative splicing of TRβ. It is possible that these isoforms may modulate T3-responsiveness in a tissue- and/or developmental stage-specific manner.

Tissue- and development-specific expression of TRs

Most studies of TR isoform expression have employed mRNA analyses rather than protein measurement (30). In general, the α and β receptor isoforms are distributed widely and exhibit overlapping patterns of expression (30,75). TRα 1 mRNA is expressed in skeletal and cardiac muscle whereas TRβ-1 mRNA is predominant in liver, kidney, and brain. Α-2 mRNA is most prevalent in brain and testis. In contrast, TRβ-2 mRNA has the most tissue-restricted expression, and is present in the anterior pituitary gland, hypothalamus, and cochlea (75-79).

 

The TRs also are expressed in specific stages during development, and are subject to regulation by hormones and other factors (78, 79). For instance, TRα-1 mRNA is expressed early whereas TRβ 1 mRNA is expressed later during embryonic brain development. In the rat pituitary gland, TH decreases TRβ 2, TRα-1, and α-2 mRNAs while slightly increasing TRβ-1 mRNA. However, in most other tissues, TH decreases TRα-1 and α-2, but not TRβ-1 mRNA. Isoform-specific knockout mice of each of the TR isoforms display distinct phenotypes (17,18). However, lack of significant TR isoform-specific gene expression was observed in cDNA microarrays of hepatic genes in TR isoform knockout mice (80). Given the apparent redundancy in TR isoform function, it is possible the different KO phenotypes may be due to absolute TR expression levels in critical tissues and developmental stages.

TRANSCRIPTIONAL REGULATION BY TRS

TH receptors bind to TH response elements (TREs) in specific target genes (Figure 3d-3, below). After binding TH, the receptor induces changes in gene expression by either increasing or decreasing the transcriptional activity of target genes. Examples of the target genes that are positively- and negatively-regulated by TH are summarized in Table 1. cDNA microarrays have been employed to study TH regulation of hepatic genes in mice, and led to the identification of a large number of novel target genes (both positively- and negatively-regulated) (81,82). These studies demonstrated that TH affected gene expression in a wide range of cellular pathways and functions, including gluconeogenesis, lipogenesis, insulin signaling, adenylate cyclase signaling, cell proliferation, and apoptosis. Although many of the TH-responsive genes were regulated directly by TRs, others were probably regulated indirectly through intermediate genes. Indirect regulation of TH-mediated transcription is suggested when the time course for induction is slow (hours) and when it is blocked by protein synthesis inhibitors. Although TH acts mainly at the level of transcription, it also can affect mRNA stability, translational efficiency, and miRNA regulation (83,84). Thus, TH acts at multiple levels to alter protein expression.

Figure 3. Mechanism of TH action via its nuclear receptor. TH is transported across plasma membrane and likely diffuses through nuclear membrane to bind to its receptor. The TH receptor (open circle) is localized almost exclusively in the nucleus where it associated with DNA as a homodimer or as a heterodimer with RXR (stippled box). The hormone-activated receptor binds to TH response elements (TREs) to alter rates of gene transcription and consequently levels of mRNA.

Table 1. Examples of Genes Positively-regulated by T3.

       
     1. Fatty acid synthetase
     2. Growth hormone
     3. Lysozyme silencer
     4. Malic enzyme
     5. Moloney leukemia virus enhancer
     6. Myelin basic protein
     7. Myosin heavy chain α
     8.Phosphoenolpyruvate carboxykinase
     9. RC3
   10. Spot 14 lipogenic enzyme
   11. Type I 5'-deiodinase
   12. Uncoupling protein

 

Table 2. Examples of Genes Negatively-regulated by T3.

 
 
     1. Epidermal growth factor receptor
     2. Myosin heavy chain β
     3. Prolactin
     4. Thyroid-stimulating hormone α
     5. Thyroid-stimulating hormone β
     6.Thyrotropin-releasing hormone
     7. Type II 5’-deiodinase
 

 

 

 

TR binding to TH response elements (TREs)

 

Detailed analyses of thyroid response elements (TREs) have led to the identification of a canonical TRE half-site sequence (1, 2,85) (Figure 3d-4). The TRE half-site is generally considered to be a hexamer (AGGTCA), but TR binding is optimal with a more extended binding site (37,88,89). Specifically, the sequence TAAGGTCA is optimal for TR binding and T3-responsiveness. However, inspection of TREs from many different target genes reveals there is a relatively low degree of sequence conservation among these elements. This finding suggests the possibility that naturally-occurring TREs may have diverged from an ideal consensus element during evolution as a means to modulate the degree of TH responsiveness.

 

TR interactions with DNA are quite different from those observed with steroid receptors, which bind to palindromic DNA sequences as homodimers. Although TR also can bind to certain TREs in vitroas a homodimer, it binds preferentially to most TREs as a heterodimer with the retinoid X receptors (RXRs) (1-3,30). The TR-RXR heterodimer binds to half-sites that are arranged in several different configurations. These include palindromic arrangements (head-to-head), direct repeats (head-to-tail), and inverted repeats (tail-to-tail). Most naturally occurring TREs are direct repeats (Figure 3d-4), typically separated by four nucleotides. The ability of TR dimers to bind to TRE’s in different configurations suggests a flexible protein structure, or the possibility that distinct protein surfaces are involved in the formation of dimers (34,39,90). Taken together, the specificity and affinity for the TR-RXR heterodimer is primarily determined by sequences within the half-site, the length of the spacer region between the half sites, and the sequence context within the spacer region.

Figure 4. Consensus thyroid response element (TRE). Studies of TRE’s in many different promoters has allowed the derivation of a "consensus" TRE comprised of a direct repeat of the hexameric sequence, AGGTCA, spaced by four nucleotides (n). Of note, there is considerable diversity in the sequences of half-sites, orientation of half-sites, and bases that form the spacers between half-sites (see text).

Although TR can interact with a wide variety of other nuclear receptors and transcriptional adaptor proteins (see below), the RXR proteins (α, β, and g) represent its most important heterodimeric partners (1-3). The RXR proteins enhance TR binding to DNA and reduce the rate of receptor dissociation from DNA (91). RXR binds to the 5’ sequence and TR binds to the 3’ sequence of TREs in which half-sites are arranged as direct repeats (92, 93). The DNA-binding domains interact with the major grooves of the half-sites on the same face of the DNA (34,92,93). The carboxy-terminal end of the TR DNA-binding domain forms an α -helical structure that interacts with the spacer region in the DNA minor groove between the TRE half-sites. Although protein-protein contacts between the RXR and TR DNA-binding domains are important for dimerization, the major sub-regions involved in dimerization reside in the carboxy-terminii of the receptors (34). The dimerization surface of the TR appears to involve residues that lie along the surfaces of helices 10 and 11. T3binding enhances the formation of TR-RXR heterodimers (94). On the other hand, T3dissociates TR-TR homodimers (95). These findings raise the possibility that T3binding might induce disruption of TR homodimers and induce the formation of TR-RXR heterodimers. The RXRs bind a stereoisomer of all trans retinoic acid, 9-cis retinoic acid (96,97). which variably alters transcriptional activity depending on the nature of the TH responsive gene (1,2). Additional studies are required to clarify the functional roles of RXRs and their ligands in TH action and interaction with other transcription factors.  Recently, Hollenberg and colleagues recentlyanalyzed the hepatic TRβcistrome of hyper- and hypothyroid mice using Chip-seq technology (98).  They found that the majority of TRβ-1 binding sites were not in the proximal promoter region but in other portions of target genes. Interestingly, by comparing the TR binding sites with previous Chip-seq data for RXRa, they found that some target genes may be regulated by TR homodimers rather TR/RXR heterodimers. Additionally, T3increased TRβ-1 binding to DNA sites that, in turn, was correlated with T3-induced gene expression. DR-4 and DR-0 motifs were significantly enriched at the DNA binding sites where T3increased or decreased in TRβ1 binding, and were associated with positive and negative transcriptional regulation by T3. Interestingly, in another study using Chip-chip methodology (99), some TH-regulated genes were identified that had little TR binding, despite the presence of putative TREs suggesting that other mechanisms such as receptor cross-talk, non-genomic effects, or indirect signaling mechanisms (see below) may be involved in regulating these genes.

TRs can have cross-talk with other nuclear hormone receptors owing to their common abilities to heterodimerize with RXRs.  TR crosstalk with peroxisome proliferator-activated receptor (PPAR) and LXR signaling via heterodimerization with RXR is a prominent example.   PPARg regulates the expression of its target genes by binding to the PPAR response element (direct repeat 1; DR1) as a heterodimer with RXR. Recently, it was shown that TRβ1 competes with PPARg for binding to DR1 as a heterodimer with RXR in vitroand in vivoto repress the transcriptional activity of PPARg (100). Since  PPARg plays a key role in lipid metabolism, carcinogenesis, and cardiovascular diseases (101.102), this mode of TR may exert some of its effects by crosstalk with PPARs. Recently, cell-based studies indicate that TRβ inhibits the activity of LXR-α transcription activity of the CYP7A1 promoter which shares a common DR-4 element with TR (103).  These studies show that TR cross-talk with other nuclear hormone receptor-mediated signaling expands TR effects beyond those target genes directly regulated by TRs (104).

 

Basal repression/Transcriptional corepressors

After binding to DNA, TR alters transcriptional activity by interacting directly or indirectly with a complex array of transcriptional cofactors. These proteins include corepressors (CoRs), coactivators (CoAs), integrators like CREB-binding protein (CBP), and general transcription factors (GTFs) (reviewed in 1-3, 31). Many of these factors have been identified by protein-protein interaction assays such as the yeast two-hybrid and glutathione-s-transferase pull down assays.

In the absence of TH, TR represses basal transcription in proportion to the amount of receptor and the affinity of receptor binding sites in positively-regulated target genes.This phenomenon also is referred to as transcriptional silencing (105-108). (Figure 3d-5, below). The addition of TH reverses basal repression and increases transcriptional activation above basal levels seen in the absence of receptor. Our understanding of the molecular mechanism for basal repression of transcription by unliganded receptor was advanced significantly by the discovery of a family of repressor proteins that bind selectively to unliganded TRs and RARs. This corepressor family includes silencing mediator for retinoid and TH receptors (SMRT) and nuclear receptor corepressor (NCoR) (105,109,110). These corepressors are 270 kD proteins that contain three transferable repression domains and two carboxy-terminal α -helical interaction domains. They are able to mediate basal repression by TR and RAR, as well as orphan members of the nuclear hormone receptor family such as rev-erbAα and chicken ovalbumin upstream transcription factor (COUP-TF). They have little or no interaction with steroid hormone receptors and therefore do not mediate basal repression by these receptors. Another protein, small ubiquitous nuclear co-repressor (SUN-CoR) enhances basal repression by TR and rev-erbA (31). This 16kD protein may form part of a co-repressor complex as it interacts with NCoR.

 

Within the interaction domains of NCoR and SMRT are consensus LXXI/HIXXXI/L sequences which resemble the LXXLL sequences that enable co-activators to interact with nuclear hormone receptors (40-42) (see below). Interestingly, these motifs allow both corepressors and co-activators to interact with similar amino acid residues on helices 3, 5, and 6 which are part of the ligand-binding pocket of TR. Differences in the length and specific sequences of the co-repressor and co-activator interaction sites coupled with the conformational changes in the LBD upon ligand binding, determine whether corepressor or coactivator binds to TR.

 

Recently, it has been shown that corepressors can form a complex with other repressors such as Sin 3 and histone deacetylases that are mammalian homologs of well-characterized yeast transcriptional repressors RPD1 and RPD3 (1-3,31). Thus, local histone deacetylation likely plays a critical role in the basal repression by unliganded TR/corepressor complex by maintaining local chromatin structure in a state that decreases basal transcription. Upon T3binding, TR undergoes a conformational change that dissociates CoRs and recruits an array of coactivators (CoAs). Thus, hormone binding relieves repression and stimulates transcription by altering receptor binding to distinct classes of cofactors. Additionally, DNA-methylation may play a role in basal repression as methyl-CpG-binding proteins can associate with a co-repressor complex containing Sin3 and histone deacetylases (111,112). This repression was relieved by the deacetylase inhibitor, trichostatin A. These findings suggest that two repression processes, DNA methylation and histone deacetylation, may be linked via methyl-CpG-binding proteins.

 

The fact that TR alters the level of gene transcription in both the absence and presence of T3has important implications for TH physiology. At low hormone concentrations, such as hypothyroidism, the unliganded receptor is predicted to repress transcription rather than function as an inactive, passive receptor. In some respects, this model is borne out by targeted inactivation of the TRα and TRβ genes. The phenotype of these double knockout mice are, for the most part, much less pronounced than the clinical features of congenital hypothyroidism (113,114). Thus, basal repression of transcription may explain why absence of receptor has less deleterious effects than absence of hormone (80,113,114).

Figure 5. TH receptor-mediated transcriptional silencing and activation. (A) Positively regulated genes. In the absence of hormone, the unliganded TH receptor represses or "silences" transcription in a process that involves TR interactions with a corepressor complex. Binding of T3 releases corepressors, relieving silencing and inducing the recruitment of coactivators that mediate transcriptional stimulation. (B) Negatively regulated genes. In the absence of hormone, the unliganded receptor activates transcription in a process that involves corepressors. Addition of TH dissociates corepressors and recruits coactivators. In the case of negatively regulated genes, this T3-mediated exchange of corepressors and coactivators inhibits transcription.

Figure 6. Role of corepressors and coactivators in the control of T3-regulated genes. In the absence of T3, the RXR-TR heterodimer recruits corepressors (CoR), which in turn, assemble additional components of a repressor complex that includes histone deacetylase (HDAC). Deacetylation of histones induce transcriptional repression. In the presence of T3, the corepressor complex dissociates and coactivators (CoA) bind to TR. The coactivator complex can include steroid receptor co-activators (SRCs)/p160, CREB-binding protein (CBP), p300/CBP associated factor (P/CAF), and proteins with histone acetyltransferase (HAT) activity. Vitamin D receptor interacting protein/TR associated protein (DRIP/TRAP) complex can also interact with liganded TR, and may cycle with SRC/p160 complex. The general transcription factors (GTFs) are also indicated.

Transcriptional activation/Coactivators

A large and growing number of co-factors have been shown to interact with liganded nuclear hormone receptors and enhance their transcriptional activation. These include: steroid receptor coactivator 1 (SRC1); SRC2/transcriptional intermediary factor 2 (TIF2) / glucocorticoid receptor interacting protein 1 (GRIP1); SRC3/ amplified in breast cancer 1 (AIB1)/ receptor associated coactivator 3 (RAC3)/ p300/CBP cointegrator associated protein (p/CIP)/ nuclear receptor coactivator (ACTR)/ thyroid receptor activator molecule 1 (TRAM 1); peroxisome proliferator activated protein binding protein (PBP); TR accesory proteins (TRAPs) /vitamin D receptor interacting proteins (DRIPs); p300/CBP associated factor (p/CAF), and cAMP response element binding protein (CREB) binding protein (CBP)/ p300. among others (reviewed elsewhere (1-3,31).

 

At present, the precise roles of all these putative coactivators are not known; however, it appears that there are at least two major complexes involved in ligand-dependent transcriptional activation: the steroid receptor co-activator (SRC) complex and the vitamin D receptor interacting protein/TR associated protein (DRIP/TRAP complex) (Fig. 3d-6). SRCs (SRC-1,SRC-2, and SRC-3) are 160 kD proteins that associate with nuclear hormone receptors, including TRs, and enhance their ligand-dependent transcription (115-117). SRCs also interact with the CREB-binding protein (CBP), the co-activator for cAMP-stimulated transcription as well as the related protein, p300, which interacts with the viral co-activator E1A (118-121). Recent studies also have shown that CBP/p300 can interact with PCAF (p300/CBP-associated factor), the mammalian homolog of a yeast transcriptional activator, general control nonrepressed protein 5, GCN5. Like GCN5, PCAF has intrinisic histone acetyltransferase activity (HAT) activity. Both PCAF and CBP interact with TBP associated factors (TAFs) and RNA pol II. Thus, PCAF and CBP possess dual functional roles both as adaptors of nuclear receptors to the basal transcriptional machinery as well as enzymes that can alter chromatin structure by histone acetyl transferase (HAT) activity. SRC‑1 and CBP may coordinate with TRs to synergize further the actions of TH, and also allow for the convergence of plasma membrane and nuclear hormone receptor signaling pathways in the cell.

 

The DRIP/TRAP complex also interacts with liganded VDRs and TRs (122-125). However, none of the subunits are members of the SRC family or their associated proteins. Instead, several DRIP/TRAP components are mammalian homologs of the yeast Mediator complex, which associates with RNA Pol II. Thus, TR recruits DRIP/TRAP complex which, in turn, may recruit or stabilize RNA Pol II holoenzyme via their shared subunits. It is noteworthy that DRIP/TRAP complex does not appear to have intrinisic HAT activity. Recent chromatin immunoprecipitation assays of proteins bound to hormone response elements (HREs), suggest that there may be a sequential, possibly cyclical recruitment, of co-activator complexes to hormone response elements by liganded nuclear hormone receptors (126-129). Studies of co-activator recruitment to TH-regulated genes showed distinct temporal patterns of recruitment. Last, other co-factors such as SW1/Snf and BRG-1 may be involved in early chromatin remodeling before the co-activator complexes are recruited to the TREs (130,131).

Negative regulation by TRs

In contrast to positively-regulated target genes, negatively-regulated genes can be stimulated in the absence of TH and repressed by TH (Figure 3d-5, above). Regulation of TRH and the TSH α  and β -subunit genes have been studied most extensively as models of negatively-regulated genes. From a physiological perspective, negative-regulation of these genes represents a critical aspect of feedback control of the TH axis. The T3-responsive regions of these negatively- regulated genes have been localized to the proximal promoter regions (132-134). However, TR binding to putative TREs in these promoters is relatively weak in comparison to the binding sites in positively-regulated genes.

 

There are several different potential mechanisms for negative regulation by TH. Negative regulation may involve receptor interference with the actions of other transcription factors or with the basal transcription apparatus (135,136). For instance, TR can inhibit the activity of AP-1, a heterodimeric transcription factor composed of Jun and Fos. T3-mediated repression of the prolactin promoter has been proposed to occur by preventing AP-1 binding (137). The TR also interacts with other classes of transcription factors, including NF-1, Oct-1, Sp-1, p53, Pit-1, CTCF, and GATA (138-144). By binding to these, or other positive transcription factors, the TH receptor may be able to inhibit gene expression by protein-protein interactions. Negative regulation may also occur by TR directly binding to DNA. A negative TRE from the TSHβ gene resides in an exon downstream of the start site of transcription (134) raising the possibility that it occludes the formation of a transcription complex. (Figure 3d-6, above) Additionally, liganded TRs may potentially recruit positive cofactors off DNA (squelching), which in turn, could lead to decreased transcription of target genes.

 

Transcriptional CoRs and CoAs, or even novel co-factors, may be involved in the control of negatively regulated genes. In contrast to the basal repression by unliganded TR in the case of positively regulated genes, CoRs cause basal activation of the TSH and TRH genes (132-134,145,146). CoAs also play an apparently paradoxical role in T3-dependent repression of negatively regulated genes (146,147). Moreover, both SRC-1 knockout mice and knockin mice which express a TRβ mutant with a mutation in the helix 12 region (that interacts with CoAs) have defective negative regulation of TSH (148,149).  Interestingly, histone acetylation can be increased in the T3- mediated negative regulation of TSHawhereas it is decreased in regulation of TSHβ and TRH (150,151).

 

Epigenetic modifications by TRs

Transcriptional regulation by TRs is a multistep process involving: (1) association of TRs with regulatory sites in the genome (usually within the targe gene promoters) in the context of chromatin, (2) ligand-dependent recruitment and function of coregulators to modify chromatin and thereby regulating RNA Pol II recruitment to the target genes, and (3)co-valent modifications of histones to alter chromatin structure, recruit RNA pol II complex, and to mediate transcription. In particular, the site-specific acetylation of histone tails induces local relaxation of chromatin, which enhances the binding of some transcriptional regulators and facilitates the recruitment and functioning of the general transcriptional machinery. Recent studies have demonstarted that thyroid hormone-positively regulated target genes may have distinct patterns of coactivator recruitment and histone acetylation that may enable highly specific regulation (129). However the epigenetic changes associated with negetively regulated gene seems to be much more complex. For instance, histone acetylation of H3K9 and H3K18 sites, two modifications usually associated with transcriptional activation, occur in negative regulation of TSHapromoter. T3also caused the release of a corepressor complex composed of histone deacetylase 3 (HDAC3), transducin b-like protein 1, and nuclear receptor coprepressor (NCoR)/ silencing mediator for retinoic and thyroid hormone receptor from TSHapromoter in chromatin immunoprecipitation assays. These findings demonstrate the critical role of NCoR/HDAC3 complex in negative regulation of TSHagene expression and show that similar complexes and overlapping epigenetic modifications can participate in both negative and positive transcriptional regulation (150).  Of note, histone deacetylation has been observed in T3-mediated negative regulation of several target genes (150-152).  Moreover, abberant histone modification at the TRH and TSHagenes has been implicated in the inappropriate TSH secretion observed in resistance to thyroid hormone (RTH) syndrome (150,151)). Other coregulators may be involved in T3-mediated regulation as RIP140, a coregulator that can decrease transcription by some nuclear hormone receptors, mediated T3repression of Crabp1 gene via chromatin remodeling during adipocyte differentiation (153). Interestingly, the use of HDAC inhibitors to counteract the effects of basal repression of target genes have restored some transcriptional activity in hypothyroidism associated with RTH syndrome and hypothyroidism (154,155). Although nuclear CoRs play a prominent role in T3 nuclear action (156,157), NCoR-independent signaling may account for basal repression by unliganded TRs for a significant number of target genes (158).

Another mechanism for T3-mediated epigenetic signaling is regulation of small non-coding microRNAs. MicroRNAs act as negative regulators of gene expression by inhibiting the translation or promoting the degradation of target mRNAs. Since individual microRNAs often regulate the expression of multiple target genes with related functions, modulating the expression of a single microRNA can, in principle, influence an entire gene network and thereby modify complex disease phenotypes (159). Thyroid hormone have been shown to regulate the levels of microRNA pair miR-206/miR-133b in human skeletal muscles (160), miR 208a in heart (161), miR21 and miR181d in liver (162,163).  These miRNAs regulate important cellular events by TH such as differentiation, contractility, and metabolism.  MicroRNAs thus are a novel mechanism for thyroid hormone signaling which may regulate mRNA levels of target genes in which TRs are not recruited to their promoters or directly affect their transcription (164).   Last, it recently has been reported that miRNA 27a can modulate the expression of target gene, b-MHC in cardiac myocytes by decreasing TRb mRNA expression (165) and multiple miRNAs may also regulate TRb expression in papillary thyroid cancer (166) suggesting direction regulation of TR expression may be another mechanism for modulating target gene expression by TH.

 

Novel indirect pathways for TH action

It has been assumed that early transcriptional activation of target genes are mediated by direct transcriptional effects by TRs owing to their abilities to bind to TREs and recruit co-activators (167,168). Previous studies have suggested that TH may have non-genomic signaling activation that may result in rapid transcriptional changes (3, see below). However, several groups have shown that TH can activate SIRT-1 activity by a TR-dependent process, that in turn, can lead to deacetylation and activation of transcription factors such as PGC1a and FoxO1a.  These findings raise the possibility that TRs can activate some target genes without TREs through activating other transcription factors (169).  On the other hand, TRs can interact with SIRT-1 directly so it is possible that it can recruite deacetylase activity that can act on transcription factors as well as modulate transcription through histone modification (170-72).

 

Thyroid Hormone Receptors and Carcinogenesis

There are many reports providing evidence that reduced TR expression and/or alterations in TH levels are common events in human cancer (173, 174). These alterations include loss of heterozygosity, gene rearrangements, promoter methylation, aberrant splicing and point mutations (173,175). Tumors, including lung, breast, head and neck, melanoma, renal, uterine, ovarian and testicular tumors, present high frequencies of somatic deletions and mutations in  both TR alpha/beta loci (176-178). Aberrant TRs have also been found in more than 70% of human hepatocellular carcinomas The tendency for TR expression to disappear as malignancies progress suggests that TR can act as a tumor suppressor in human cancers; therefore, loss of expression and/or function of this receptor could result in cell transformation and tumor development (179). In fact, TR overexpression in hepatoma cell lines shows repression of various tumor promoting genes such as PTTG1 (180),  and activation of  anti-tumorogenic TGF-beta. However certain mutant TRs like TRbPV/PVmey even enhance tumor growth by non-genomicaly activating beta-catenin and PI3K pathways (181,182). Last, it recently has been reported that miRNAs can downregulate the expression of dio 1 in renal cell carcinoma, and TRbin papillary thyroid, carcinoma.  Clarifying the molecular mechanisms by which TRs influence tumor progression and elucidating the epigenetic modifications ofT3target genes in cancers would perhaps lead to a better understanding of the treatment regime in humans.

 

 

Recently, targeted gene inactivation or knockout (KO) of TR isoforms, and “knockin” of mutant TRs to their native TR genomic locii have provided new information on the mechanisms of TH action (16,17). The ability to disrupt TR genes by targeted mutagenesis has been particularly challenging given there is more than one gene encoding TRs, multiple splicing variants (TRα-1, α-2, TRβ-1, TRβ-2), and an additional transcript (Rev-erbA) derived from the opposite strand of the TRα gene (12,16,17). Two TRα knockout mouse lines have been generated that display different phenotypes (175, 176). It is likely this difference is due to the different sites in the TRα gene locus used for homologous recombination to generate the knockout mice. The TRα gene is complex as it encodes TRα-1, α-2 (which cannot bind T3), and rev-erbA (generated from the opposite strand encoding TRα) (12, 16, 17). KO mice in which both TRα-1 and α-2 were deleted (TRα -/-) had a more severe phenotype with hypothyroidism, intestinal malformation, growth retardation, and early death shortly after weaning (175). T3injection prevented the early death of pups. KO mice that lacked only TRα-1 (TRα-1-/-) had a milder phenotype with decreased body temperature and prolonged QT intervals on electrocardiograms (183). The phenotypic effects of the loss of TRα 1 are relatively mild (184-185). Unexpectedly, there is no evidence of resistance to TH, as occurs with the TRβknockout. Disruption of the TRα-1 causes lower heart rates (19% reduced) and prolonged QRS and QT durations. These cardiac effects persist after hormone replacement. No changes were found in the levels of known TH-responsive genes in the heart (e.g., sarcoplasmic Ca2+ ATPase, Na+-K+ATPase, β-adrenergic receptors). The bradycardic effect of the TRα-1 knockout may result from alterations in the sympathetic or parasympathetic nervous systems or it could result from an intrinsic defect in cardiac myocytes. The TRα-1-deficient mice also have a 0.5 oC reduction in body temperature that is independent of TH levels. The mice have normal amounts of brown adipose tissue.

 

Samarut and co-workers have reported generation of short TRα isoforms from intronic transcriptional start sites which have dominant negative activity on TR function (73), and it is likely these short TRα isoforms are responsible for the more severe phenotype of the TRα -/-mice. In this connection, TRα KO mice which did not express either TRα-1 and α-2 (TRαo/o), had a milder phenotype than TRα -/-mice which expressed only the short TRα isoforms (186). Interestingly, TH stimulation of some target genes was increased, perhaps due to the absence of α-2 which inhibits normal TR-mediated transcription (57,58).

 

Targeted disruption of the TRβ locus created a mouse deficient in both TRβ-1 and TRβ-2 (16,17). These mice had elevated circulating TSH and T4levels, thyroid hyperplasia, as well as hearing defects (187,188). These findings are similar to the index patients with resistance to TH who were later shown to have homozygous deletion of TRβ (4,152). Thus, the mouse model appears to faithfully reproduce some of the features seen in humans with resistance to TH who are lack TRβ or express a dominant negative mutant TRβ (189). TRβ-2-selective knockout mice also have been generated and exhibited elevated levels of TH and TSH suggesting TRβ-2 plays the major role in regulating TSH (190). TRβ-2-selective knockout mice also have abnormal color discrimination and suggest TRβ-2 may play a role in cone development of the retina (191).

 

The relatively mild phenotypes of the TRα-1 and TRβ KO mice suggest the two isoforms have redundant roles in the transcriptional regulation of many target genes. In this connection, microarray studies of TRα and TRβ KO mice showed similar gene regulation profiles in the absence and presence of T3in liver (82).  Recently, a study employing TRα or TRbreceptor over-expressing cell lines also showed that both these receptor isoforms mostly share a common gene repertoire but with varying degrees of induction or repression of target genes (192).

When both TR isoforms were abolished, the resultant double knockout mice (TRα 1-/-TRβ-/-) were surprisingly viable (113, 114). Thus, the absence of TRs is compatible with life. These mice had markedly elevated T4, T3, and TSH as well as large goiters. They also showed decreased growth, fertility, heart rate as well as bone density and development. Interestingly comparison of cDNA microarrays of double KO and hypothyroid mice showed only partial overlap of their gene regulation profiles, confirming the observation that the absence of receptor can give a different phenotype than lack of hormone. It is likely that basal transcription occurs even in the absence of receptor whereas basal repression of target genes occurs in the absence of hormone.  When the phenotypes of TRa, TRb, and double KO are compared, it is apparent that each isoform may have isoform-specific function, perhaps in part due to different expression patterns of the isoforms as well as gene-specific actions by each isoform (114).

 

Cheng and colleagues have generated a “knock-in” mouse model in which a mutant TRβ from a patient with RTH (PV) was introduced into the endogenous TRβ gene locus (193). These mice have a phenotype similar to patients with RTH, as the heterozygous mice showed elevated serum T4and TSH, mild goiter, hypercholesterolemia, impaired weight gain, and abnormal bone development. Homozygous mice had markedly elevated serum T4and TSH, and a much more severe phenotype than heterozygous mice. Wondisford and colleagues also have generated a “knock-in” mouse that expresses mutant TRβ (194). These mice had abnormal cerebellar development and function, and learning deficits. These latter studies suggest that expression of mutant TRβ under the control of endogenous TRβ promoter produces many of the clinical features of RTH in mice. This same group also recently developed a knock-in of a mutant TRβ that cannot bind DNA. This model should be useful in distinguishing signaling and developmental patterns due to protein-protein interactions of TRs (as well as non-genomic pathways) from those that require TR binding to TREs of target genes (195). Knockin mice harboring a TRamutant at the same site as the TRβPVmutant gene decreased white adipose tissue (WAT) and liver mass (196). In contrast, TRβPV markedly induced hepatosteatosis and mass of liver but had little effect on white adipose tissue. The expression of lipogenic genes was decreased in white adipose tissue and liver of TRaPVmice whereas it was increased in liver and normal in TRβPVmice. A recent study showed that the phenotype of impaired adipogenesis can be restored by crossing with mice expressing a mutant Ncor1 allele (Ncor1(ΔID) mice) that cannot recruit the TR (197). These findings support the notion that the phenotypes in the TRaPVmutant mice, and perhaps some patients with RTH with mutant TRa, may be due to aberrant repression of target gene (18, 198, 199).

NONGENOMIC PATHWAYS REGULATED BY TH

 

There is increasing evidence for non-genomic effects by TH (3) in addition to the transcriptional effects mediated by nuclear TRs. There is continuous shuttling of a small amount of TRs between the cytoplasm and nucleus (200), so non-genomic effects may be mediated by cytoplasmic TRs (see below). Recently, a TRavariant from alternative translation was shown to be palmitoylated and associated with the plasma membrane (201). TH binding to this receptor led to increased intracellular calcium, nitrous oxide, and cyclic guanine monophosphate (cGMP), which in turn activated PKGII, Src, ERK, and Akt signaling pathways.  Another recent study suggests that non-DNA-binding TRs that cannot stimulate transcription may have “non-canonical” thyroid hormone signaling to regulate important physiological effects such as serum glucose and triglyceride  levels, body temperature, and heart rate. (202).  However, it appears that many non-genomic effects by TH are  likely mediated by cellular binding proteins other than TRs. Evidence supporting this notion comes from the rapid time course of some TH effects (thus precluding transcription and protein synthesis), utilization of membrane-signaling pathways such as kinases or calmodulin, lack of dependence on the presence of nuclear TRs, and structure-activity correlations by TH analogs that are different than those observed for nuclear TRs (3). Several non-nuclear sites for TH binding have been identified in various cell systems although their functional significances are not well characterized. Some of these include: plasma membrane associated T3transporters, actin, calcium ATPase, adenylate cyclase, and glucose transporters; an endoplasmic reticulum associated protein, prolyl hydroxylase; and monomeric pyruvate kinase  (3, 203-207). A useful guideline that describes transcriptional and non-transcriptional signaling via TR and non-TR mechanisms recently has been published (208)

 

TH also has profound effects on mitochondrial activity and cellular energy state. A 43 kD protein has been described in mitochondria which also could bind to TREs and could be recognized by antibodies against the TRα ligand-binding domain (209). Recently, it has been shown that TRβ can interact with the p85 subunit of PI3K and activate the PI3K-Akt/PKB signaling cascade; thus, the small subpopulation of cytosolic TRβmay be involved in cell signaling (210). This PI3K activation by T3leads to both direct and indirect effects on the transcription of several genes involved in glucose metabolism (210, 211,) and provides a mechanism for cross-talk between TH and cell signaling pathways.

 

Recently, integrin α-Vβ-3, has been identified as a plasma membrane TH-binding site (212). Previously, T4, but not T3, was shown to promote actin polymerization and integrin interaction with laminin in neural cells (213). Additionally, both T4 and T3activated mitogen-activated protein kinase (MAPK) activity, and led, among other events, to phosphorylation of TRβ (210). Using a chick chorioallantoic membrane (CAM) system, Davis et al. showed that both T4 and T3stimulated angiogenesis (214). Since integrin α-Vβ-3 is involved in angiogenesis, T4and T3binding to it was examined, and T4was found to bind to integrin α-Vβ-3 with high affinity. Tetraiodothyroacetic acid (tetrac) and antibodies against laminin blocked T4binding (213). Moreover, siRNAs against the integrin α-V or β-3 subunits blocked MAPK activation by TH. These findings suggest that TH activates the APK cascade and stimulates angiogenesis via TH binding to integrin α Vβ 3. Additionally, thyroid hormone non-genomically suppresses Src thereby stimulating osteocalcin expression in primary mouse calvarial osteoblasts (215). A direct physical interaction of TRbPV with cellular proteins, namely the regulatory subunit of the phosphatidylinositol 3-kinase (p85alpha), the pituitary tumor transforming gene (PTTG) and beta-catenin, that are critically involved in cell proliferation, motility, migration, angiogenesis and metastasis suggest a novel mode of non-genomic action, whereby mutant TR isoform acts as an oncogene in thyroid carcinogenesis (216).

TH ANALOGS, METABOLITES, AND ANTAGONISTS

 

Several tissue-specific and TR isoform-specific compounds have been developed as potential treatments for hypercholesterolemia, obesity, and heart failure. An early prototypical compound was 3,5-dibromo-3-pyridazinone-L-thyronine (L-940901) that bound preferentially to the TRs in the liver over those in the heart (217). Although the relative affinity of this compound for the respective TR isoforms has not been reported, the selective action of L-940901 is likely due to tissue-specific uptake of the compound. Interestingly, mice treated with L-940901 had decreased serum cholesterol levels without cardiotoxicity. Recently, several other TH analogs have been described that have isoform-selective affinity for TRβ  compared to TRα (218-220). Since TRs in the liver are approximately 90% TRβ whereas those in the heart are mostly TRα, these isoform-selective compounds may serve as novel agents to lower serum cholesterol with minimal cardiotoxicity. N-[3,5-dimethyl-4-(4’-hydroxy-3’isopropylphenoxy)-phenyl]-oxamic acid (CGS 23425), 3,5-dimethyl-4(4’-hydroxy-3’-isopropylbenzyl)-phenoxy) acetic acid (GC-1), and 3,5-dichloro-4[(4-hydroxy-3-isoopropylphenoxy)phenyl] acetic acid (KB-141) all have been reported to lower total serum cholesterol and LDL-cholesterol (205-209). CGS 23425 also increases LDL receptor expression in HepG2 cells. Additionally, these compounds can increase serum apoA1 levels; however, the total serum high density lipoprotein (HDL) cholesterol level does not changes or may even decrease. In this connection, GC-1 decreased serum HDL; increased expression of HDL receptor, SR-B1; stimulated the activity of cholesterol 7α hydroxylase; and increased fecal excretion of bile acids in treated mice (221). Thus, GC-1 regulates important steps in the reverse cholesterol transport pathway (221). Recently, KB141 was shown to be a potential treatment for obesity by decreasing body weight via stimulation of metabolic rate and oxygen consumption (222). The TR agonist MB07811, which is converted to an active metabolite in the liver, has proven to be effective in reducing hepatic steatosis in rodents (223). The TRb-specific compound, KB215 recently was shown to be effective in decreasing LDL cholesterol, apoliprotein B, triglycerides, and Lp(a) in humans (224)when used in combination with statins.  These findings suggest that TH analogs may be useful in the treatment of a wide range of metabolic disorders (225).

 

TH analogs and derivatives also bind specifically to proteins other than TRs, and are involved in non-genomic cell signaling pathways, Thyronamines (3-T1AM, T0AM) are endogenous compounds derived from L-thyroxine or its intermediate metabolites. Activities of intestinal deiodinases and ornithine decarboxylase generate 3-T1AM (226). Significantly, this compound bound poorly to nuclear TRs. T1AM has interesting physiological actions as it produced a rapid drop in body temperature and heart rate when injected intraperitoneally in mice. T1AM also decreased cardiac output in an ex vivo working heart model. Although 3-T1AM have a weak affinity towards classical nuclear TH receptors a number of putative receptors, binding sites, and cellular target molecules mediating actions of 3-T1AM have been proposed. Among those are members of the trace amine associated-receptor family (TAR1), the adrenergic receptor ADRα2a, and the thermosensitive transient receptor potential melastatin 8 channel (226). Preclinical studies  using animal models are in progress, and more stable receptor-selective agonistic and antagonistic analogues of 3-T1AM are now being synthesiszed exerting marked cryogenic, metabolic, cardiac and central actions and represents a key lead compound linking endocrine, metabolic, and neuroscience research to advance development of new drugs (226).

 

TH can increase cardiac performance by increasing cardiac contractility and decreasing systemic vascular resistance (2); however, TH excess also can cause cardiotoxicity. 3,5-diiodothyropropionic acid (DITPA) is a TH-related compound with low metabolic activity and low affinity for nuclear TRs (Kd 10-7M). DITPA was able to increase cardiac contractility and peripheral circulation without significant effects on heart rate in animal studies (227). Moreover, DITPA improved hemodynamic performance in animal models of congestive heart failure after myocardial infarction. Patients with heart failure treated with DITPA showed significant improvement in systolic cardiac index and systemic vascular resistance in preliminary studies (227). Thus DITPA or similar compounds may represent a novel class of drugs for the treatment of heart failure.

 

Recently, the naturally occurring analogs, 3,5,3'-triiodothyroacetic (TRIAC) and 3,5,3',5'-tetraiodothyroacetic TETRAC) acids decreased heat-induced albumin fibrillation suggesting they may have a protective effect against amyloid formation (228). Additionally, tetraiodothyroacetic acid (tetrac) caused radiosensitization of GL261 glioma cells (229).  The mechanisms for these effects are not understood at this time but likely involve non-genomic effects as TRIAC and TETRAC have weak binding affinity for TRs.  Similarly, 3,5-diiodothyronine (T2) has also shown favorable effects in rodent models of fatty liver diseases (230,231).

SUMMARY

 

We have learned much about molecular mechanisms of nuclear TH action during the past 25 years. In particular, the identification and characterization of TRs, their heterodimeric partners, corepressors, coactivators, and TREs, generation of TR knockout mice, and discovering non-genomic pathways have provided new insight into TH action. It is expected that new information will be obtained from microarray and proteomic studies, structural biology approaches, and in vitro transcriptional systems. Such information should provide an even better understanding of the mechanisms of disease caused by abnormal circulating TH and/or altered intracellular TH levels, and provide targets for the development useful therapeutic agents for not only TH-related conditions but also for metabolic derangements such as hypercholesterolemia, non-alcoholic fatty liver disease, and obesity.

 

BIBLIOGRAPHY

  1. Yen PM: Physiological and molecular basis of TH action. Physiol Rev 81:1097-1142, 2001.
  2. Brent, G: Mechanisms of thyroid hormone action. J Clin Invest. 2012 122(9): 3035–3043, 2012
  3. Cheng SY, Leonard JL, Davis PJ. Molecular Aspects of Thyroid Hormone Actions. Endocr Rev. 31: 139–170, 2010.
  4. Zulewski H, Muller B, Exer PM, et al: Estimation of tissue hypothyroidism by a new clinical score: Evaluation of patients with various grades of hypothyroidism and controls. J Clin Endocrinol Metab 82:771-776, 1997.
  5. Dumitrescu AM, Refetoff SThe syndromes of reduced sensitivity to thyroid hormone. Biochim Biophys Acta.1830:3987-4003, 2013
  6. Al-Adsani H, Hoffer LJ, Silva JE: Resting energy expenditure is sensitive to small dose changes in patients on chornic TH replacement. J Clin Endocrinol Metab 82:1118-1125, 1997.
  7. Magnus-Levey A: Ueber den respiratorischen gaswechsel unter dem einfluss der thyroidea sowie unter verschiedenen pathologische zustand. Berl Klin Wschr 32:650, 1895.
  8. Cheng SY, Leonard JL, Davis PJ. Molecular aspects of thyroid hormone actions.Endocr Rev.31:139-70, 2010.
  9. Tata JR, Widnell CC: Ribonucleic acid synthesis during the early action of TH. Biochem J 98:604-620, 1966.
  10. Schadlow AR, Surks MI, Schwartz HL, et al: Specific triiodothyronine binding sites in the anterior pituitary of the rat. Science 176:1252-1254, 1972.
  11. Samuels HH, Tsai JS, Casanova J, et al: TH action: in vitro characterization of solubilized nuclear receptors from rat liver and cultured GH1 cells. J Clin Invest 54:853-865, 1974.
  12. Koerner D, Schwartz HL, Surks MI, et al: Binding of selected iodothyronine analogues to receptor sites of isolated rat hepatic nuclei. High correlation between structural requirements for nuclear binding and biological activity. J Biol Chem 250:6417-6423, 1975.
  13. Samuels HH, Stanley F, Casanova J: Relationship of receptor affinity to the modulation of TH nuclear receptor levels and growth hormone synthesis by L- triiodothyronine and iodothyronine analogues in cultured GH1 cells. J Clin Invest 63:1229-1240, 1979.
  14. Schwartz HL, Trence D, Oppenheimer JH, et al: Distribution and metabolism of L- and D-triiodothyronine (T3) in the rat: preferential accumulation of L-T3 by hepatic and cardiac nuclei as a probable explanation of the differential biologic potency of T3 enantiomers. Endocrinology 113:1236-1243, 1983.
  15. Sap J, Munoz A, Damm K, et al: The c-erb-A protein is a high-affinity receptor for TH. Nature 324:635-640, 1986.
  16. Weinberger C, Thompson CC, Ong ES, et al: The c-erb-A gene encodes a TH receptor. Nature 324:641-646, 1986.
  17. Forrest D Vennstrom B: Functions of TH receptors in mice. Thyroid 10: 41-52, 2000.
  18. Flamant F, Samarut J. TH receptors: lessons from knockout and knock-in mutant mice. Trends Endocrinol Metab. 2003 14:85-90.
  19. Zavacki AM, Larsen PR: RTHα, a newly recognized phenotype of the resistance to thyroid hormone (RTH) syndrome in patients with THRA gene mutations.J Clin Endocrinol Metab. 98:2684-6, 2013
  20. Yoh SM, Privalsky ML. Molecular analysis of human resistance to thyroid hormone syndrome. Methods in Molecular Biology. 202:129–152, 2002
  21. Bianco AC. Minireview: cracking the metabolic code for thyroid hormone signaling.Endocrinology. 152:3306-11, 2011.
  22. Dentice M, Marsili A, Zavacki A, Larsen PR, Salvatore D. The deiodinases and the control of intracellular thyroid hormone signaling during cellular differentiation.

Biochim Biophys Acta. 1830:3937-45, 2913.

  1. Silva JE, Dick TE, Larsen PR: The contribution of local tissue thyroxine monodeiodination to the nuclear 3,5,3'-triiodothyronine in pituitary, liver, and kidney of euthyroid rats. Endocrinology 103:1196-1207, 1978.
  2. Larsen PR, Frumess RD: Comparison of the biological effects of thyroxine and triiodothyronine in the rat. Endocrinology 100:980-988, 1977.
  3. Maia AL, Kim BW, Huang SA, Harney JW, Larsen PR Type iodothyronine deiodinase is the major source of plasma T3 in euthyroid humans. J Clin Invest 115:2524-33, 2005.

26.van der Deure WM, Peeters RP, Visser TJMolecular aspects of thyroid hormone transporters, including MCT8, MCT10, and OATPs, and the effects of genetic variation in these transporters.  J Mol Endocrinol.44:1-11, 2010.

  1. Jansen J, Friesema EC, Milici C, Visser TJ Thyroid hormone transporters in health and disease. Thyroid 15:757-68, 2005
  2. Dumitrescu AM, Liao XH, Best TB, Brockmann K, Refetoff S A novel syndrome combining thyroid and neurological abnormalities is associated with mutations in a monocarboxylate transporter gene. Am J Hum Genet 74:168-75, 2004.
  3. Rastinejad F, Perlmann T, Evans RM, et al: Structural determinants of nuclear receptor assembly on DNA direct repeats. Nature 375:203-211, 1995.
  4. Brent GA. Mechanisms of thyroid hormone action. J Clin Invest. 122:3035-43, 2012.
  5. Zhang J, Lazar MA: The mechanism of action of THs. Annu Rev Physiol. 62:439-466, 2000.
  6. Thompson CC, Weinberger C, Lebo R, et al: Identification of a novel TH receptor expressed in the mammalian central nervous system. Science 237:1610-1614, 1987.
  7. Drabkin H, Kao FT, Hartz J, et al: Localization of human ERBA2 to the 3p22----3p24.1 region of chromosome 3 and variable deletion in small cell lung cancer. Proc Natl Acad Sci U S A 85:9258-9262, 1988.
  8. Mader S, Kumar V, de VH, et al: Three amino acids of the oestrogen receptor are essential to its ability to distinguish an oestrogen from a glucocorticoid-responsive element. Nature 338:271-274, 1989.
  9. Umesono K, Evans RM: Determinants of target gene specificity for steroid/TH receptors. Cell 57:1139-1146, 1989.
  10. Rastinejad F, Perlmann T, Evans RM, et al: Structural determinants of nuclear receptor assembly on DNA direct repeats. Nature 375:203-211, 1995.
  11. Yen PM, Ikeda M, Wilcox EC, et al. 1994 Half-site arrangement of hybrid glucocorticoid and thyroid hormone response elements specifies thyroid hormone receptor complex binding to DNA and transcriptional activity. J Biol Chem 269:12704-12709.
  12. Nelson CC, Faris JS, Hendy SC, et al: Functional analysis of the amino acids in the DNA recognition alpha- helix of the human TH receptor. Mol Endocrinol 7:1185-1195, 1993.
  13. Wagner RL, Apriletti JW, McGrath ME, et al: A structural role for hormone in the TH receptor. Nature 378:690-697, 1995.
  14. Hu X, Lazar MA: The CoRNR motif controls the recruitment of corepressors by nuclear hormone receptors. Nature 402: 93-6, 1999.
  15. Nagy L, Kao HY, Love JD, et al: Mechanism of corepressor binding and release from nuclear hormone receptors. Genes Dev 13: 3209-16, 1999.
  16. Perissi, V., L. M. Staszewski, E. M. McInerney, et al: Molecular determinants of nuclear receptor-corepressor interaction. Genes Dev 13: 3198-208, 1999.
  17. Feng, W., R. C. Ribeiro, R. L. Wagner, et al: Hormone-dependent coactivator binding to a hydrophobic cleft on nuclear receptors. Science 280: 1747-9, 1998.
  18. Lazar MA, Hodin RA, Darling DS, et al: Identification of a rat c-erbAa-related protein which binds deoxyribonucleic acid but does not bind TH. Mol Endocrinol 2:893-901, 1988.
  19. Mitsuhashi T, Tennyson GE, Nikodem VM: Alternative splicing generates messages encoding rat c-erbA proteins that do not bind TH. Proc Natl Acad Sci U S A 85:5804-5808, 1988.
  20. Izumo S, Mahdavi V: TH receptor alpha isoforms generated by alternative splicing differentially activate myosin HC gene transcription. Nature 334:539-542, 1988.
  21. Nakai A, Seino S, Sakurai A, et al: Characterization of a TH receptor expressed in human kidney and other tissues. Proc Natl Acad Sci U S A 85:2781-2785, 1988.
  22. Schueler PA, Schwartz HL, Strait KA, et al: Binding of 3,5,3'-triiodothyronine (T3) and its analogs to the in vitro translational products of c-erbA protooncogenes: differences in the affinity of the a and ß forms for the acetic acid analog and failure of the human testis and kidney products to bind T3. Mol Endocrinol 4:227-234, 1990.
  23. Lazar MA, Hodin RA, Chin WW: Human carboxy-terminal variant of a-type c-erbA inhibits trans-activation by TH receptors without binding TH. Proc Natl Acad Sci U S A 86:7771-7774, 1989.
  24. Koenig RJ, Lazar MA, Hodin RA, et al: Inhibition of TH action by a non-hormone binding c-erbA protein generated by alternative mRNA splicing. Nature 337:659-661, 1989.
  25. Rentoumis A, Chatterjee VKK, Madison LD, et al: Negative and positive transcriptional regulation by TH receptor isoforms. Mol Endocrinol 4:1522-1531, 1990.
  26. Katz D, Berrodin TJ, Lazar MA: The unique C-termini of the TH receptor variant, c-erbAa2, and TH receptor a1 mediate different DNA-binding and heterodimerization properties. Mol Endocrinol 6:805-814, 1992.
  27. Nagaya T, Jameson JL: Distinct dimerization domains provide antagonist pathways for TH receptor action. J Biol Chem 268:24278-24282, 1993
  28. Yang YZ, Burgos-Trinidad M, Wu Y, et al: TH receptor variant alpha2. Role of the ninth heptad in dna binding, heterodimerization with retinoid X receptors, and dominant negative activity. J Biol Chem 271:28235-28242, 1996
  29. Tagami T, Kopp P, Johnson W, et al: The TH receptor variant alpha2 is a weak antagonist because it is deficient in interactions with nuclear receptor corepressors. Endocrinology 139:2535-2544, 1998
  30. Hodin RA, Lazar MA, Chin WW: Differential and tissue-specific regulation of the multiple rat c- erbA messenger RNA species by TH. J Clin Invest 85:101-105, 1990.
  31. Katz D, Lazar MA: Dominant negative activity of an endogenous TH receptor variant (alpha 2) is due to competition for binding sites on target genes. J Biol Chem 268:20904-20910, 1993..
  32. Farsetti A, Lazar J, Phyillaier M, et al: Active repression by TH receptor splicing variant alpha2 requires specific regulatory elements in the context of native triiodothyronine-regulated gene promoters. Endocrinology 138:4705-4712, 1997.
  33. Liu RT, Suzuki S, Miyamoto T, et al: The dominant negative effect of TH receptor splicing variant alpha 2 does not require binding to a thyroid response element. Mol Endocrinol 9:86-95, 1995.
  34. Katz D, Reginato M, Lazar M: Functional regulation of TH receptor variant TR alpha 2 by phosphorylation. Mol Cell Biol 15:2341‑2348, 1995.
  35. Miyajima N, Horiuchi R, Shibuya Y, et al: Two erbA homologs encoding proteins with different T3 binding capacities are transcribed from opposite DNA strands of the same genetic locus. Cell 57:31-39, 1989.
  36. Lazar MA, Hodin RA, Darling DS, et al: A novel member of the thyroid/steroid hormone receptor family is encoded by the opposite strand of the rat c-erbA alpha transcriptional unit. Mol Cell Biol 9:1128-1136, 1989.
  37. Spanjaard RA, Nguyen VP, Chin WW: Rat Rev-erbA alpha, an orphan receptor related to TH receptor, binds to specific TH response elements. Molecular Endocrinology 8: 286-295, 1994.
  38. Harding HP, Lazar MA: The monomer-binding orphan receptor Rev-Erb represses transcription as a dimer on a novel direct repeat. Mol Cell Biol 15:4791-4802, 1995.
  39. Zamir I, Dawson J, Lavinsky RM, et al: Cloning and characterization of a corepressor and potential component of the nuclear hormone receptor repression complex. Proc Natl Acad Sci U S A 94:14400-14405, 1997.
  40. Munroe SH, Lazar MA: Inhibition of c-erbA mRNA splicing by a naturally occurring antisense RNA. J Biol Chem 266:22083-22086, 1991.
  41. Lazar MA, Hodin RA, Cardona G, et al: Gene expression from the c-erbA alpha/Rev-ErbA alpha genomic locus. Potential regulation of alternative splicing by opposite strand transcription. J Biol Chem 265:12859-12863, 1990.
  42. Hodin RA, Lazar MA, Wintman BI, et al: Identification of a TH receptor that is pituitary- specific. Science 244:76-79, 1989.
  43. Oberste-Berghaus CK, Zanger K, Hashimoto K, et al: TH-independent interaction between the TH receptor beta2 amino terminus and coactivators. J Biol Chem 275: 1787-92, 2000.
  44. Tomura H, Lazar J, Phyillaier M, et al: The N-terminal region (A/B) of rat TH receptors alpha 1, beta 1, but not beta 2 contains a strong TH-dependent transactivation function. Proc Natl Acad Sci U S A 92:5600-5604, 1995.
  45. Satoh T, Yamada M, Iwasaki T, et al: Negative regulation of the gene for the preprothyrotropin-releasing hormone from the mouse by TH requires additional factors in conjunction with TH receptors. J Biol Chem 271:27919-27926, 1996.
  46. Safer JD, Langlois MF, Cohen R, et al: Isoform variable action among TH receptor mutants provides insight into pituitary resistance to TH. Mol Endocrinol 11:16-26, 1997.
  47. Chassande, O., A. Fraichard, K. Gauthier, et al: Identification of transcripts initiated from an internal promoter in the c-erbA alpha locus that encode inhibitors of retinoic acid receptor-alpha and triiodothyronine receptor activities. Mol Endocrinol 11: 1278-90, 1997.
  48. Williams GR. Cloning and characterization of two novel TH receptor beta isoforms. Mol Cell Biol. 20:8329-42, 2000.
  49. Forrest D, Sjoberg M, Vennstrom B: Contrasting developmental and tissue-specific expression of alpha and beta TH receptor genes. EMBO J 9:1519-1528, 1990.
  50. Cook CB, Kakucska I, Lechan RM, et al: Expression of TH receptor ß2 in rat hypothalamus. Endocrinology 130:1077-1079, 1992.
  51. Lechan RM, Qi Y, Berrodin TJ, et al: Immunocytochemical delineation of TH receptor beta 2-like immunoreactivity in the rat central nervous system. Endocrinology 132:2461-2469, 1993.
  52. Bradley DJ, Towle HC, Young WS, 3rd: Alpha and beta TH receptor (TR) gene expression during auditory neurogenesis: evidence for TR isoform-specific transcriptional regulation in vivo. Proc Natl Acad Sci U S A 91:439-443, 1994.
  53. Oppenheimer JH, Schwartz HL: Molecular basis of TH-dependent brain development 18:462-475, 1997.
  54. Yen PM, Feng X, Flamant F, Chen Y, Walker RL, Weiss RE, Chassande O, Samarut J, Refetoff S, Meltzer PS.Status and TH Receptor (TR) Isoforms on Hepatic Gene Expression Profiles in TR Knockout Mice. EMBO reports 4:581-587, 2003.
  55. Feng X, Yuan J, Meltzer PB, Yen PM. TH regulation of hepatic genes in vivo detected by cDNA microarray. Mol Endocrinol 2000; 14: 947-955.
  56. Flores-Morales A, Gullberg H, Fernandez L, et al: Patterns of liver gene expression governed by TRbeta.Mol Endocrinol. 16:1257-68, 2002.
  57. Leedman PJ, Stein AR, Chin WW Regulated specific protein binding to a conserved region of the 3'-untranslated region of thyrotropin beta-subunit mRNA. Mol Endocrinol 9:375-87, 1995.
  58. Van Rooij E, Marshall WS, Olson EN. Toward microRNA-based therapeutics for heart disease: the sense in antisense. Circ Res. 103:919-28, 2008.
  59. RW, Subauste JS, Koenig RJ: The interplay of half-site sequence and spacing on the activity of direct repeat TH response elements. J Biol Chem 270:5238-5242, 1995.
  60. Katz RW, Koenig RJ: Nonbiased identification of DNA sequences that bind TH receptor alpha 1 with high affinity. J Biol Chem 268:19392-19397, 1993.
  61. Umesono K, Murakami KK, Thompson CC, et al: Direct repeats as selective response elements for the TH, retinoic acid, and vitamin D receptors. Cell 65:1255-1266, 1991.
  62. Näär A, Boutin J, SM L, et al: The orientation and spacing of core DNA-binding motifs dictate selective transcriptional responses to three nuclear receptors. Cell 65:1267-1279, 1991.
  63.  Mangelsdorf D, Evans R: The RXR heterodimers and orphan receptors. Cell 83:841-850, 1995.
  64. Wahlstrom GM, Sjoberg M, Andersson M, et al: Binding characteristics of the TH receptor homo- and heterodimers to consensus AGGTCA repeat motifs. Mol Endocrinol 6:1013-1022, 1992.
  65. Yen PM, Brubaker JH, Apriletti JW, Baxter JD, Chin WW 1994 Roles of T3 and DNA-binding on thyroid hormone receptor complex formation. Endocrinology 134:1075-1081
  66. Perlmann T, Rangarajan PN, Umesono K, et al: Determinants for selective RAR and TR recognition of direct repeat HREs. Genes Dev 7:1411-1422, 1993.
  67. Zechel C, Shen XQ, Chen JY, et al: The dimerization interfaces formed between the DNA binding domains of RXR, RAR and TR determine the binding specificity and polarity of the full-length receptors to direct repeats. Embo J 13:1425-1433, 1994.
  68. Collingwood TN, Butler A, Tone Y, et al: TH-mediated enhancement of heterodimer formation between TH receptor beta and retinoid X receptor. J Biol Chem 272:13060-13065, 1997.
  69. Yen PM, Darling DS, Carter RL, et al: Triiodothyronine (T3) decreases binding to DNA by T3-receptor homodimers but not receptor-auxiliary protein heterodimers. J Biol Chem 267:3565-3568, 1992.
  70. Levin AA, Sturzenbecker LJ, Kazmer S, et al: 9-cis retinoic acid stereoisomer binds and activates the nuclear receptor RXRa. Nature 355:359-361, 1992.
  71. Heyman RA, Mangelsdorf DJ, Dyck JA, et al: 9-cis retinoic acid is a high affinity ligand for the retinoid X receptor. Cell 68:397-406, 1992.
  72. Ramadoss P, Abraham BJ, Tsai L, Zhou Y, Costa-E-Sousa RH, Ye F, Bilban M, Zhao K, Hollenberg AN Novel Mechanism of Positive versus Negative Regulation by Thyroid Hormone Receptor β1 (TRβ1) Identified by Genome-wide Profiling of Binding Sites in Mouse Liver.J Biol Chem. 289:1313-28, 2014.
  73. Gagne R, Green JR, Dong H, Wade MG, YaukC. Identification of thyroidhormone receptor binding sites in developing mouse cerebellum. BMC Genomics.14:341, 2013.
  74. 100. Araki O, Ying H, Furuya F, Zhu X, Cheng SY. Thyroid hormone receptor b mutants: dominant negative regulators of peroxisome proliferator-activated receptor g action. Proc Natl Acad Sci USA 102:16251–16256, 2005.
  75. Hansen MK,Connolly TM. Nuclear receptors as drug targets in obesity, dyslipidemia and atherosclerosis. Curr Opin Investig Drugs 9:247–255, 2008.
  76. Hart CM The role of PPARa in pulmonary vascular disease. J Investig Med 56:518–521, 2008.
  77. Hashimoto K, Cohen RN, Yamada M, Markan KR, Monden T, Satoh T, Mori M, Wondisford FE. Crosstalk between thyroid hormone receptor and liver X receptor regulatory pathways is revealed in a thyroid hormone resistance mouse model. J Biol Chem 281:295–302, 2006.
  78. Yan-Yun Liu, Gregory A. Brent. Thyroid Hormone Crosstalk with Nuclear Receptor Signaling in Metabolic Regulation. Trends Endocrinol Metab. Trends Endocrinol Metab. 21: 166–173, 2010.
  79. Torchia J, Glass C, Rosenfeld MG: Co-activators and co-repressors in the integration of transcriptional responses. Curr Opin Cell Biol. 10:373-383, 1998.
  80. Brent GA, Dunn MK, Harney JW, et al: TH aporeceptor represses T3-inducible promoters and blocks activity of the retinoic acid receptor. New Biol 1:329-336, 1989.
  81. Damm K, Thompson CC, Evans RM: Protein encoded by v-erbA functions as a thyroid-hormone receptor antagonist. Nature 339:593-597, 1989.
  82. LazarMA. Thyroidhormoneaction: a binding contract. J Clin Invest.112:497-9, 2003.
  83. Mottis A, Mouchiroud L, Auwerx J. Emerging roles of the corepressors NCoR1 and SMRT in homeostasis.Genes Dev. 27:819-35, 2013.
  84. Horlein AJ, Naar AM, Heinzel T, et al: Ligand-independent repression by the TH receptor mediated by a nuclear receptor co-repressor. Nature 377:397-404, 1995.
  85. Nan X, Ng H, Johnson CA et al: Transcriptional repression by the methyl-CpG-binding protein MeCP2 involves a histone deacetylase complex. Nature 393: 386-9, 1998.
  86. Wade PA, Gegonne A, Jones PL, et al: Mi-2 complex couples DNA methylation to chromatin remodelling and histone deacetylation. Nat Genet 23: 62-6, 1999.
  87. Gothe S, Wang Z, Ng L, et al: Mice devoid of all known TH receptors are viable but exhibit disorders of the pituitary-thyroid axis, growth, and bone maturation. Genes Dev 13: 1329-41, 1999.
  88. Gauthier K, Chassande O, Plateroti M, et al: Different functions for the TH receptors TRalpha and TRbeta in the control of TH production and post-natal development. Embo J 18: 623-31, 1999.
  89. Onate SA, Tsai SY, Tsai MJ, et al: Sequence and characterization of a coactivator for the steroid hormone receptor superfamily. Science 270:1354-1357, 1995.
  90. Voegel JJ, Heine MJS, Zechel C, et al: TIF2, a 160 kDa transcriptional mediator for the ligand-dependent activation function AF-2 of nuclear receptors. EMBO J 15:3667-3675, 1996.
  91. Hong H, Kohli K, Trivedi A, et al: GRIP1, a novel mouse protein that serves as a transcrptional coactivator in yeast for the hormone binding domains of steroid receptors. Proc Natl Acad Sci USA 93:4948-4952, 1996.
  92. Yang X-J, Ogryzko VV, Nishikawa J, et al: A p300/CBP-associated factor that competes with the adenoviral oncoprotein E1A. Nature 319-324, 1996.
  93. Kwok RP, Lundblad JR, Chrivia JC, et al: Nuclear protein CBP is a coactivator for the transcription factor CREB. Nature 370:223-226, 1994.
  94. Eckner R, Ewen ME, Newsome D, et al: Molecular cloning and functional analysis of the adenovirus E1A- associated 300-kD protein (p300) reveals a protein with properties of a transcriptional adaptor. Genes Dev 8:869-884, 1994.
  95. Torchia J, Rose DW, Inostrova J, et al: The transcriptional coactivator p/CIP binds CBP and mediates nuclear-receptor function.Nature 387:677-684, 1997.
  96. Fondell JD, Guermah M, Malik S, et al: TH receptor-associated proteins and general positive cofactors mediate TH receptor function in the absence of the TATA box-binding protein-associated factors of TFIID. Proc Natl Acad Sci U S A 96:1959-1964, 1999.
  97. Rachez C, Suldan Z, Ward J, et al: A novel protein complex that interacts with the vitamin D3 receptor in a ligand-dependent manner and enhances VDR transactivation in a cell- free system. Genes Dev 12:1787-1800, 1998.
  98. Ito M, Roeder RG: The TRAP/SMCC/Mediator complex and TH receptor function. Trends Endocrinol Metab. 12:127-134, 2001.
  99. Rachez C, Freedman, LP: Mediator complexes and transcription. Curr Opin Cell Biol. 13:274-280, 2001.
  100. Sharma D, Fondell JD: Ordered recruitment of histone acetyltransferases and the TRAP/Mediator complex to TH-responsive promoters in vivo. Proc Natl Acad Sci U S A. 99:7934-7939, 2002.
  101. Shang Y, Hu X, DiRenzo J, et al: Cofactor dynamics and sufficiency in estrogen receptor-regulated transcription. Cell. 103:843-852, 2000.
  102. Reid G, Hubner MR, Metivier R,et al: Cyclic, proteasome-mediated turnover of unliganded and liganded ERalpha on responsive promoters is an integral feature of estrogen signaling. Mol Cell 11: 695-707, 2003.
  103. Liu Y, Xia X, Fondell JD, Yen PM. Thyroid hormone-regulated target genes have distinct patterns of coactivator recruitment and histone acetylation. Mol Endocrinol 20:483-90, 2006.
  104. DiRenzo J, Shang Y, Phelan M, et al. BRG-1 is recruited to estrogen-responsive promoters and cooperates with factors involved in histone acetylation. Mol Cell Biol 20:7541-9, 2000.
  105. Nagaich AK, Walker DA, Wolford R, Hager GL. Rapid periodic binding and displacement of the glucocorticoid receptor during chromatin remodeling. Mol Cell 14:163-74, 2004.
  106. Hollenberg AN, Monden T, Flynn TR, et al: The human thyrotropin-releasing hormone gene is regulated by TH through two distinct classes of negative TH response elements. Mol Endocrinol 9:540-550, 1995.
  107. Chatterjee VKK, Lee JK, Rentoumis A, et al: Negative regulation of the thyroid-stimulating hormone alpha gene by TH: receptor interaction adjacent to the TATA box. Proc Natl Acad Sci U S A 86:9114-9118, 1989.
  108. Bodenner DL, Mroczynski MA, Weintraub BD, et al: A detailed functional and structural analysis of a major TH inhibitory element in the human thyrotropin beta-subunit gene. J Biol Chem 266:21666-21673, 1991.
  109. Zhang XK, Wills KN, Husmann M, et al: Novel pathway for TH receptor action through interaction with jun and fos oncogene activities. Mol Cell Biol 11:6016-6025, 1991.
  110. Lopez G, Schaufele F, Webb P, et al: Positive and negative modulation of Jun action by TH receptor at a unique AP1 site. Mol Cell Biol. 1993 13:3042-9.
  111. Pernasetti F, Caccavelli L, Van de Weerdt C, et al: TH inhibits the human prolactin gene promoter by interfering with activating protein-1 and estrogen stimulations. Mol Endocrinol 11:986-996, 1997.
  112. Tansey WP, Catanzaro DF: Sp1 and TH receptor differentially activate expression of human growth hormone and chorionic somatomammotropin genes. J Biol Chem 266:9805-9813, 1991.
  113. Schaufele F, West BL, Reudelhuber TL: Overlapping Pit-1 and Sp1 binding sites are both essential to full rat growth hormone gene promoter activity despite mutually exclusive Pit-1 and Sp1 binding. J Biol Chem 265:17189-17196, 1990.
  114. Schaufele F, West BL, Baxter JD: Synergistic activation of the rat growth hormone promoter by Pit-1 and the TH receptor. Mol Endocrinol 6:656-665, 1992.
  115. Barrera-Hernandez G, Zhan Q, Wong R, et al: TH receptor is a negative regulator in p53-mediated signaling pathways. DNA Cell Biol 17:743-750, 1998.
  116. Qi JS, Yuan Y, Desai-Yajnik V, et al: Regulation of the mdm2 oncogene by TH receptor. Mol Cell Biol 19:864-872, 1999.
  117. Lutz M, Burke LJ, LeFevre P, et al: TH-regulated enhancer blocking: cooperation of CTCF and TH receptor The EMBO Journal 22: 1579-1587, 2003.
  118. Matsushita A, Sasaki S, Kashiwabara Y, Nagayama K, Ohba K, Iwaki H, Misawa H, Ishizuka K, Nakamura H. Essential role of GATA2 in the negative regulation of thyrotropin beta gene by thyroid hormone and its receptors. Mol Endocrinol. 21:865-84, 2007.
  119. Wang D, Xia X, Liu Y, Oetting A, Walker RL, Zhu Y, Meltzer P, Cole PA, Shi YB, Yen PM. Negative regulation of TSHalpha target gene by thyroid hormone involves histone acetylation and corepressor complex dissociation. Mol Endocrinol. 23:600-9, 2009.
  120. Tagami T, Madison LD, Nagaya T, et al: Nuclear receptor corepressors activate rather than suppress basal transcription of genes that are negatively regulated by TH. Mol Cell Biol 17:2642-2648, 1997.
  121. Tagami T, Gu WX, Peairs PT, et al: A novel natural mutation in the TH receptor defines a dual functional domain that exchanges nuclear receptor corepressors and coactivators. Mol Endocrinol 12:1888-1902, 1998.
  122. Weiss RE, Xu J, Ning G, Pohlenz J, O'Malley BW, Refetoff S. Mice deficient in the steroid receptor co-activator 1 (SRC-1) are resistant to thyroid hormone. Embo J 18:1900-4, 1999.
  123. Ortiga-Carvalho TM, Shibusawa N, Nikrodhanond A, et al. Negative regulation by thyroid hormone receptor requires an intact coactivator-binding surface. J Clin Invest 115:2517-23, 2005.
  124. Wang D, Xia X, Weiss RE,Refetoff S,Yen PM.Distinct and histone-specific modifications mediate positive versus negative transcriptional regulation of TSHalpha promoter.PLoS One.5:e9853, 2010
  125. Umezawa R, Yamada M, Horiguchi K, Ishii S, Hashimoto K, Okada S, Satoh T, Mori M.Aberrant histone modifications at the thyrotropin-releasing hormone gene in resistance to thyroid hormone: analysis of F455S mutant thyroid hormone receptor.Endocrinology. 150:3425-32, 2009.
  126. Sasaki S, Lesoon-Wood LA, Dey A, et al: Ligand-induced recruitment of a histone deacetylase in the negative-feedback regulation of the thyrotropin beta gene. Embo J 18: 5389-98, 1999.
  127. Park SW, HuangW-H, Persaud SD, WeiLNRIP140in thyroid hormone-repression and chromatin remodeling of Crabp1 gene during adipocyte differentiation. Nucleic Acids Research, Vol. 37, No. 21 7085-7094, 2009.
  128. Kim DW, Park JW, Willingham MC, Cheng SY. A histone deacetylase inhibitor improves hypothyroidism caused by a TRα1 mutant.Hum Mol Genet. 23, 2651-2664, doi:10.1093/hmg/ddt660 (2014).

 

  1. Kumar, P., Mohan, V., Sinha, R. A., Chagtoo, M. & Godbole, M. M. Histone deacetylase inhibition reduces hypothyroidism-induced neurodevelopmental defects in rats. The Journal of endocrinology 227, 83-92, doi:10.1530/joe-15-0168 (2015).

 

  1. Astapova, I. Role of co-regulators in metabolic and transcriptional actions of thyroid hormone. Journal of molecular endocrinology 56, 73-97, doi:10.1530/jme-15-0246 (2016).

 

  1. Vella KR, Hollenberg AN.The actions of thyroid hormone signaling in the nucleus.Mol Cell Endocrinol. 2017 Dec 15;458:127-135.

 

  1. Mendoza, A. et al. NCoR1-independent mechanism plays a role in the action of the unliganded thyroid hormone receptor. Proceedings of the National Academy of Sciences of the United States of America 114, E8458-e8467, doi:10.1073/pnas.1706917114 (2017).

 

  1. Van Rooij E, Marshall WS, Olson EN. Toward microRNA-based therapeutics for heart disease: the sense in antisense. Circ Res. 103:919-28, 2008.
  2. Visser WE, Heemstra KA, Swagemakers SM, Ozgür Z, Corssmit EP, Burggraaf J, van Ijcken WF, van der Spek PJ, Smit JW, Visser TJ. Physiological thyroid hormone levels regulate numerous skeletal muscle transcripts. J Clin Endocrinol Metab. 94:3487-96, 2009
  3. 161. Callis TE, Pandya K, Seok HY, Tang RH, Tatsuguchi M, Huang ZP, Chen JF, Deng Z, Gunn B, Shumate J, Willis MS, Selzman CH, Wang DZ. MicroRNA-208a is a regulator of cardiac hypertrophy and conduction in mice. J Clin Invest. 119:2772-86, 2009.
  4. Yap CS, Sinha RA, Ota S, Katsuki M, Yen PM. Thyroid hormone negatively regulates CDX2 and SOAT2 mRNA expression via induction of miRNA-181d in hepatic cells.Biochem Biophys Res Commun. 440:635-9, 2013.
  5. Huang YH, Lin YH, Chi HC, Liao CH, Liao CJ, Wu SM, Chen CY, Tseng YH, Tsai CY, Lin SY, Hung YT, Wang CJ, Lin CD, Lin KH.Thyroid hormone regulation of miR-21 enhances migration and invasion of hepatoma.Cancer Res. 73:2505-17, 2013.
  6. Dong H, Paquette M, Williams A, Zoeller RT, Wade M, Yauk C. Thyroid hormone may regulate mRNA abundance in liver by acting on microRNAs.PLoS One. 13;5:e12136, 2010.
  7. Hitoo Nishi, Koh Ono, Takahiro Horie, et al. MicroRNA-27a Regulates Beta Cardiac Myosin Heavy Chain Gene Expression by Targeting Thyroid Hormone Receptor β1 in Neonatal Rat Ventricular Myocytes Mol Cell Biol. 31: 744–755, 2011
  8. Jazdzewski K, Boguslawska J, Jendrzejewski J, Liyanarachchi S, Pachucki J, Wardyn KA, Nauman A, de la Chapelle A. Thyroid hormone receptor beta (THRB) is a major target gene for microRNAs deregulated in papillary thyroid carcinoma (PTC).J Clin Endocrinol Metab. 96:E546-53, 2011.
  9. Ramadoss, P. et al. Novel mechanism of positive versus negative regulation by thyroid hormone receptor beta1 (TRbeta1) identified by genome-wide profiling of binding sites in mouse liver. The Journal of biological chemistry 289, 1313-1328, doi:10.1074/jbc.M113.521450 (2014).
  10. Grontved, L. et al. Transcriptional activation by the thyroid hormone receptor through ligand-dependent receptor recruitment and chromatin remodelling. Nature communications 6, 7048, doi:10.1038/ncomms8048 (2015).
  11. Thakran S, Sharma P, Attia RR, Hori RT, Deng X, Elam MB, Park EA. Role of sirtuin 1 in the regulation of hepatic gene expression by thyroid hormone. J Biol Chem.288:807-18, 2013 .
  12. Singh BK, Sinha RA, Zhou J, Xie SY, You SH, Gauthier K, Yen PM. FoxO1 deacetylation regulates thyroid hormone-induced transcription of key hepatic gluconeogenic genes.J Biol Chem. 288(42):30365-72, 2013.
  13. Singh, B. K. et al. Hepatic FOXO1 Target Genes Are Co-regulated by Thyroid Hormone via RICTOR Protein Deacetylation and MTORC2-AKT Protein Inhibition. The Journal of biological chemistry 291, 198-214, doi:10.1074/jbc.M115.668673 (2016).

172.Suh JH, Sieglaff DH, Zhang A, Xia X, Cvoro A, Winnier GE, WebbP. SIRT1is a direct coactivator of thyroid hormone receptor β1 with gene-specific actions. PLoS One.8(7):e70097, 2013.

  1. Aranda A, Martínez-Iglesias O, Ruiz-Llorente L, García-Carpizo V, Zambrano A. Thyroid receptor: roles in cancer. Trends Endocrinol Metab. 20:318-24, 2009.
  2. Frau, C. et al. Local hypothyroidism favors the progression of preneoplastic lesions to hepatocellular carcinoma in rats. Hepatology (Baltimore, Md.) 61, 249-259, doi:10.1002/hep.27399 (2015)
  3. Bonamy, G.M. and Allison, L.A. Oncogenic conversion of thyroid hormone receptor by altered nuclear transport. Nucl. Recept. Signal. 4, e008, 2006.
  4. Gonzalez-Sancho, J.M. et al. Thyroid hormone receptors/THR genes in human cancer. Cancer Lett. 192, 121–132, 2003.
  5. Conde, I. et al. Influence of thyroid hormone receptors on breast cancer cell proliferation. Ann. Oncol. 17, 60–64, 2006.
  6. Horkko, T.T. et al. Thyroid hormone receptor beta1 in normal colon and colorectal cancer-association with differentiation, polypoid growth type and K-ras mutations. Int. J. Cancer 118, 1653–1659. 2006.

179 Chan, I.H. and Privalsky, M.L. Thyroid hormone receptors mutated in liver cancer function as distorted antimorphs. Oncogene 25, 3576–3588, 2006.

  1. Chen, R.N. et al. Thyroid hormone receptors suppress pituitary tumor transforming gene 1 activity in hepatoma. Cancer Res. 68, 1697–1706, 2008.
  2. Guigon CJ, Zhao L, Lu C, Willingham MC, Cheng SY. Regulation of β-catenin by a novel nongenomic action of thyroid hormone β receptor. Mol Cell Biol 28:4598 –4608, 2008.
  3. Furuya, F. et al. Activation of phosphatidylinositol 3-kinase signaling by a mutant thyroid hormone beta receptor. Proc. Natl. Acad. Sci. U. S. A. 103, 1780–1785, 2006.
  4. Wikstrom L, Johansson C, Salto C, et al: Abnormal heart rate and body temperature in mice lacking TH receptor alpha 1. Embo J 17, 455-461, 1998.
  5. Fraichard A, Chassande O, Plateroti, M et al: The T3R alpha gene encoding a TH receptor is essential for post-natal development and TH production. Embo J 16, 4412-4420, 1997.
  6. Johansson C, Vennstrom B, Thoren P: Evidence that decreased heart rate in TH receptor-alpha1- deficient mice is an intrinsic defect. Am J Physiol 275:R640-646, 1998.

186.Macchia, P.E., Takeuchi, Y., Kawai, T., Cua, K., Gauthier, K., Chassande, O., Seo, H., Hayashi, Y., Samarut, J., Murata, Y., Weiss, R.E. and Refetoff, S. Increased sensitivity to TH in mice with complete deficiency of TH receptor alpha. Proc Natl Acad Sci U S A 98, 349-354, 2001.

  1. Forrest D, Hanebuth E, Smeyne RJ, et al: Recessive resistance to TH in mice lacking TH receptor beta: evidence for tissue-specific modulation of receptor function. Embo J 15:3006-3015, 1996.
  2. Weiss RE, Forrest D, Pohlenz J, et al: Thyrotropin regulation by TH in TH receptor beta-deficient mice. Endocrinology 138:3624-3629, 1997.
  3. Refetoff S, DeWind LT, DeGroot LJ: Familial syndrome combining deaf-mutism, stippled epiphyses, goiter and abnormally high PBI: possible target organ refractoriness to TH. J Clin Endocrinol Metab 27:279-294, 1967.
  4. Abel ED, Boers ME, Pazos-Moura, C, et al: Divergent roles for TH receptor beta isoforms in the endocrine axis and auditory system. J Clin Invest 104, 291-300, 1999.
  5. Ng L, Hurley JB, Dierks, B, et al: A TH receptor that is required for the development of green cone photoreceptors. Nat Genet 27, 94-98, 2001.
  6. Chan IH, Privalsky ML.Isoform-specific transcriptional activity of overlapping target genes that respond to thyroid hormone receptors alpha1 and beta1.Mol Endocrinol. 2009 23:1758-75.
  7. Kaneshige M, Kaneshige K, Zhu X, et al: Mice with a targeted mutation in the TH beta receptor gene exhibit impaired growth and resistance to TH. Proc Natl Acad Sci U S A 97, 13209-13214, 2000.
  8. Hashimoto K, Curty FH, Borges PP, et al: An unliganded TH receptor causes severe neurological dysfunction. Proc Natl Acad Sci U S A 98, 3998-4003, 2001.
  9. Shibusawa N, Hashimoto K, Nikrodhanond AA, et al: TH action in the absence of TH receptor DNA-binding in vivo. J Clin Invest. 112: 497-9, 2003.
  10. Araki O, Ying H, Zhu XG, Willingham MC, Cheng SY. Distinct dysregulation of lipid metabolism by unliganded thyroid hormone receptor isoforms.Mol Endocrinol. 2):308-15, 2009.
  11. Fozzatti L, Kim DW, Park JW, Willingham MC, Hollenberg AN, Cheng SY. Nuclear receptor corepressor (NCOR1) regulates in vivo actions of a mutated thyroid hormone receptor α.Proc Natl Acad Sci U S A. 110(19):7850-5, 2013

 

  1. Bochukova E, Schoenmakers N, Agostini M, et al.A mutation in the thyroid hormone receptor gene. N Engl J Med. 366:243–249, 2012.
  2. vanMullemA, van Heerebeek R, Chrysis D, et al. Clinical phenotype and mutant TR_1. N Engl J Med. 2012;366:1451–1453, 2012.
  3. Baumann CT, Maruvada P, Hager GL, Yen PM. Nuclear cytoplasmic shuttling by thyroid hormone receptors. multiple protein interactions are required for nuclear retention. J Biol Chem 276:11237-45, 2001.
  4. Kalyanaraman, H. et al. Nongenomic thyroid hormone signaling occurs through a plasma membrane-localized receptor. Science signaling 7, ra48, 2014.
  5. Hones GS, Rakov H, Logan J et al. Noncanonical thyroid hormone signaling mediates cardiometabolic effects in vivo. Proc Nat Acad Sci 114: E11323-E11332, 2017
  6. Cheng SY, Gong QH, Parkison C, et al. The nucleotide sequence of a human cellular thyroid hormone binding protein present in endoplasmic reticulum. Journal of Biological Chemistry 262:11221-11227, 1987.
  7. Kato H, Fukuda T, Parkison C, McPhie P, Cheng SY. Cytoplasmic thyroid hormone-binding protein is a monomer of pyruvate kinase. Proceedings of the National Academy of Science 86:7681-7685, 1990.
  8. Sterling K, Brenner MA. Thyroid hormone action: effect of triiodothyronine on mitochondrial adenine nucleotide translocase in vivo and in vitro. Metabolism 44:193-199, 1995.

206.Leonard JL, Farwell AP. Thyroid hormone-regulated actin polymerization in brain. Thyroid 7:147-151, 1997

  1. Davis, P. J., Goglia, F. & Leonard, J. L. Nongenomic actions of thyroid hormone. Nature reviews. Endocrinology 12, 111-121, doi:10.1038/nrendo.2015.205 (2016).
  2. Flamant F, Cheng SY, Hollenberg AN et al. Thyroid HormoneSignaling Pathways: Time for a More Precise Nomenclature.Endocrinology. 158:2052-2057, 2017
  3. Wrutniak C, Cassar-Malek I, Marchal S, et al. A 43 kD protein related to c-erb A alpha 1 is located in the mitochondrial matrix of rat liver. Journal of Biological Chemistry 270:16347-16354, 1995.
  4. Cao X, Kambe F, Moeller LC, Refetoff S, Seo H Thyroid hormone induces rapid activation of Akt/protein kinase B-mammalian target of rapamycin-p70S6K cascade through phosphatidylinositol 3-kinase in human fibroblasts. Mol Endocrinol 19:102-12, 2005.
  5. Moeller LC, Dumitrescu AM, Refetoff S. Cytosolic action of thyroid hormone leads to induction of hypoxia-inducible factor-1alpha and glycolytic genes. Mol Endocrinol 19:2955-63, 2005.
  6. Bergh JJ, Lin HY, Lansing L, et al. Integrin alphaVbeta3 contains a cell surface receptor site for thyroid hormone that is linked to activation of mitogen-activated protein kinase and induction of angiogenesis. Endocrinology 146:2864-71, 2005.
  7. Farwell AP, Tranter MP, Leonard JL. Thyroxine-dependent regulation of int3grin-laminin interactions in astrocytes. Endocrinology 136:3909-15, 1995.
  8. Davis PJ, Davis FB, Cody V. Membrane receptors mediating thyroid hormone action. Trends Endocrinol Metab 16:429-35, 2005.
  9. Asai S, Cao X, Yamauchi M, Funahashi K, Ishiguro N, Kambe F. Thyroid homone non-genomically suppresses Src thereby stimulating osteocalcin expression in primary mouse calvarial osteoblasts. Biochem Biophys Res Commun. 387:92-6, 2009.
  10. Guigon CJ, Cheng SY. Novel non-genomic signaling of thyroid hormone receptors in thyroid carcinogenesis. Mol Cell Endocrinol. 308:63-9, 2009.
  11. Underwood AH, Emmett JC, Ellis D, et al. A thyromimetic that decreases plasma cholesterol levels without increasing cardiac activity. Nature 324:425-9, 1986
  12. Morkin E, Ladenson P, Goldman S, Adamson C Thyroid hormone analogs for treatment of hypercholesterolemia and heart failure: past, present and future prospects. J Mol Cell Cardiol 37:1137-46, 2004.
  13. Taylor AH, Stephan ZF, Steele RE, Wong NC Beneficial effects of a novel thyromimetic on lipoprotein metabolism. Mol Pharmacol 52:542-7, 1997.
  14. Chiellini G, Apriletti JW, Yoshihara HA, Baxter JD, Ribeiro RC, Scanlan TS A high-affinity subtype-selective agonist ligand for the thyroid hormone receptor. Chem Biol 5:299-306, 1998.
  15. Johansson L, Rudling M, Scanlan TS, et al. 2005 Selective thyroid receptor modulation by GC-1 reduces serum lipids and stimulates steps of reverse cholesterol transport in euthyroid mice. Proc Natl Acad Sci U S A 102:10297-302.

222 Grover GJ, Mellstrom K, Ye L, et al. Selective thyroid hormone receptor-beta activation: a strategy for reduction of weight, cholesterol, and lipoprotein (a) with reduced cardiovascular liability. Proc Natl Acad Sci U S A 100:10067-72, 2003.

  1. Cable EE, Finn PD, Stebbins JW, Hou J, Ito BR, van Poelje PD, Linemeyer DL, Erion MD. Reduction of hepatic steatosis in rats and mice after treatment with a liver-targeted thyroid hormone receptor agonist. Hepatology. 49:407-17, 2009.
  2. Ladenson PW, Kristensen JD,Ridgway EC,Olsson AG,Carlsson B,Klein I,Baxter JD,Angelin B.Use of the thyroid hormone analogue eprotirome in statin-treated dyslipidemia. N Engl J Med.362:906-16, 2010.
  3. ShoemakerTJ, Kono T, Mariash CN, Evans-Molina C. Endocr Pract.Thyroid hormoneanalogues for the treatment of metabolic disorders: new potential for unmet clinical needs? 18:954-64, 2012.
  4. Hoefig CS, Zucchi R, Köhrle J. Thyronamines and Derivatives: Physiological Relevance, Pharmacological Actions, and Future Research Directions. Thyroid. Dec;26(12):1656-1673, 2016.
  5. Morkin E, Pennock GD, Spooner PH, Bahl JJ, Goldman S. Clinical and experimental studies on the use of 3,5-diiodothyropropionic acid, a thyroid hormone analogue, in heart failure. Thyroid 12:527-33176, 2002.
  6. Cortez LM, Farías RN, Chehín RN. Protective effect of 3,5,3'-triiodothyroacetic and 3,5,3',5'-tetraiodothyroacetic acids on serum albumin fibrillation. Eur Biophys J. 38:857-63, 2009.
  7. Hercbergs A, Davis PJ, Davis FB, Ciesielski MJ, Leith JT. Radiosensitization of GL261 glioma cells by tetraiodothyroacetic acid (tetrac). Cell Cycle. 8:2586-91, 2009.
  8. Iannucci, L. F. et al. Metabolomic analysis shows differential hepatic effects of T2 and T3 in rats after short-term feeding with high fat diet. Scientific reports 7, 2023, 2017.
  9. Sinha RA, Singh BK, Yen PM.Direct effects of thyroid hormones on hepatic lipid metabolism.Nat Rev Endocrinol. May;14(5):259-269, 2018.

Prostate Cancer Detection

ABSTRACT

Prostate cancer is the second most common cause of male cancer deaths in Western countries. However, one of the most contentious topics in medicine continues to be whether testing for this very common tumor is in the best interests of individual patients. Although there is a spectrum of progression rates for this tumor, in most instances, prostate cancer replicates and spreads slowly. As this tumor is uncommonly diagnosed before the age of 40 years and the likelihood of clinical detection increases as men age, most patients have comorbidities when diagnosed with prostate cancer. For this reason and because there are not insignificant potential disadvantages with the detection process and its consequences, it is important to determine whether the benefits of detection are likely to be greater than the unwanted effects of leaving a possible prostate cancer undiagnosed. In this Endotext chapter, the likelihood of a detectable prostate cancer being present is placed in context of patients’ ages and co-morbidies before detailing the tests currently used in clinical practice, together with their limitations. For complete coverage of all related areas of Endocrinology, please visit our on-line FREE web-text, WWW.ENDOTEXT.ORG.

INTRODUCTION AND BACKGROUND

Prostate Cancer is an increasingly common diagnosis in Western societies with over 240 000 diagnoses made in the US (1), as well as 196,200 in Asia and 417,100 in Europe each year (2).  There is a wide range in the incidence of prostate cancer across the globe with the highest rates in developed countries, being more than four-fold higher than less developed regions for a slightly lower mortality (3),  although non-Westernized societies are changing as reported recently in relation to the Asia-Pacific region (4)(Figure 1). These differences are likely multifactorial, including genetic, environmental, detection, and reporting differences. As reported for 2012, Australia and New Zealand now has had the highest age-standardized incidence (111.6) and cumulative risk (13.6% by age 75) of prostate cancer in the world, with a high incidence observed in Northern America (97.2) and Western Europe (94.9).

Figure 1: Prostate Cancer Incidence Rates for Select Registries, 2000–2004 (5)

Mortality rates vary from country to country as well (6-7)with prostate cancer following lung and bowel cancers in Europe and Australia in terms of mortality rates. Worldwide, Caribbean and African populations display the highest prostate cancer mortality rates (Figure 2) (3). This disparity is also likely multifactorial and additionally includes factors related to treatment availability and practices.

Figure 2: Prostate Cancer Age-Standardized Mortality Rates for Selected Registries, 2000–2006 (5)

Despite advances in prevention and early detection, refinements in surgical technique and improvements in radiotherapy and chemotherapy, the ability to cure many patients remains elusive. However, mortality rates are changing albeit slowly as illustrated in blue below for Australia. A 2013 report by the Australian Institute of Health and Welfarepredicts that by 2020 only 26 out of 100,000 Australian men will die from the disease compared with 34 in 1982 (Figure 3) (8).

 

This phenomenon is not peculiar to Australia. Baade et al reviewed international trends in prostate cancer mortality and reported significant reductions in prostate-cancer mortality in the UK, USA, Austria, Canada, Italy, France, Germany, Australia and Spain with downward trends in the Netherlands, Ireland and Sweden (9). This has subsequently been observed by others (4,10).

 

Earlier detection of this disease, as a consequence of the introduction of the prostate specific antigen (PSA) blood test, has been acknowledged by the NCI as one factor contributing to lowering the mortality rate over the past few years (11-14). The use of PSA testing has been estimated to provide a diagnostic lead-time of up to 10 years (15-19). In the mid to late 1980s only one third of prostate cancers were diagnosed at curable stages compared with today when 80% are staged clinically as organ-confined and potentially curable (20-22). Unfortunately, however, even when the tumor is thought to be localized, up to 25% of men have non-localized disease which declares itself subsequently (23).

Figure 3: Panel A – Incidence (solid line) during 1982 – 2014 in Australia demonstrating a rise after widespread availability of PSA testing with a dip after the prostate cancer backlog was addressed: mortality (dashed line) has been falling slowly since the mid-1990s. Panel B – 5-year relative survival from prostate cancer, 1984–1988 to 2009–2013 in Australia demonstrated a reciprocal improvement since the mid-1990s https://prostate-cancer.canceraustralia.gov.au/statistics

Since curative treatments are limited to localized tumors (11-12,15,24), extending effective but non-invasive treatments to include both primary and secondary lesions remains a major goal and challenge. Once prostate cancer metastasizes, apart from causing loss of life, the toll it exacts is often considerable with regard to morbidity from both the disease itself and administered therapies.

 

As a result of increasing numbers of men having their prostate cancers diagnosed earlier, more patients are now eligible for treatment with curative intent.  Improved surgical and radiation-based treatments have been developed so that the prognosis of a man diagnosed today with prostate cancer is better than ever before.

 

 

ANATOMY AND PHYSIOLOGY

 

The word "prostate", originally derived from the Greek prohistani which means "to stand in front of," has been attributed to Herophilus of Alexandria who used the term in 355 BC to describe the small organ located in front of the bladder (25). The prostate gland is a small firm structure, about the size of a chestnut, located below the bladder and in front of the rectum (Figure 4). The urethra, the channel through which urine is voided, passes from the bladder through the prostate and penis (Figure 5).

Figure 4: The Normal Prostate and its Relationship to Other Pelvic Structures

The primary function of the prostate gland, which contracts with ejaculation, is to provide enzymes to maintain the fluid nature of seminal fluid and to nourish sperm as they pass through the prostatic and penile urethra to outside the body.

Figure 5: Zonal Anatomy of the Prostate (sagittal depiction)
1 = peripheral zone - the zone where most cancers originate
2 = central zone – zone in which middle lobe develops
3 = transition zone – zone in which BPH ‘lateral lobes’ form
4 = anterior zone
B = bladder
U = urethra

NATURAL HISTORY OF PROSTATE CANCER

 

Traditionally, prostate cancer was considered to be a disease of "older men." As such, it was generally accepted that "men never died fromprostate cancer, they died of other conditions with prostate cancer."  Consequently, treatment was conservative and directed toward palliation and management of any debilitating and painful sequelae. In addition, diagnosis from histopathology from a biopsy was generally made after palpating a rock-hard and nodular prostate on digital rectal exam [DRE] or by symptoms and signs of primary or secondary tumors, such as urinary obstruction, back pain, nerve root or, less commonly, spinal cord compression. In a large majority of cases, tumors had already disseminated at the time of diagnosis and, therefore, were incurable. It was in the mid-1980s, with the introduction of the PSA blood test that prostate cancer began to be diagnosed earlier and in younger men.

 

Prostate cancer is usually slow in its development and in the majority of cases, slow to progress as is illustrated in Table 1 below from Surveillance Epidemiology and End Results (SEER) registry: SEER collects and publishes cancer incidence and survival data from population-based cancer registries covering approximately 28% of the population of the United States(1).

 

If autopsy findings are an indication, premalignant and inapparent tumors are very common with one United States study indicating that, of 249 cases examined, 70% of the prostates with the premalignant condition high grade prostate intra-epithelial neoplasia (HGPIN) harbored adenocarcinoma, whereas the frequency of cancerin prostates without HGPIN was 24%. HGPIN was encountered in 0, 5, 10, 41 and 63% of men in the 3rd, 4th, 5th and 7th decades, respectively. The corresponding figures for invasive carcinoma were 2, 29, 32, 55 and 64% respectively (26).

Although methods of diagnosis and treatment of localized disease have become well-entrenched, they are beginning to change. However, both early detection through PSA screening and the management of prostate cancer remain controversial. The tumor has a variable biologic course, the traditional biopsy approach is invasive, costly and clinical staging of tumors is imprecise. Furthermore, there are significant limitations in prediction of the clinical outcome of patients with both organ-confined and extra-prostatic disease - not to mention the morbidity associated with all currently established treatments. It is sobering to muse that, were the unwanted effects of diagnosis and treatment insignificant, the dilemma of whether or not to diagnose and treat would not be issues.

 

COMPETING MORBIDITIES AND LIFE EXPECTANCY: COMPARISONS

 

The likelihood of men dying from causes other than from prostate cancer increases with ageing because of competing mortalities (as indicated by Figure 6 below), in particular cardiovascular and cerebrovascular diseases (Figure 6 below): the fact that most prostate cancers progress slowly compared with other cancers needs to be considered in terms of life expectancy from competing causes of death.  Life expectancy has been reported to be increasing for Australian men, recently estimated to be 80.4 years from birth, the 7thhighest worldwide, and 84.5 years at age 65(27). Calculation of life expectancy is difficult; however, use of statistically calculated “life tables”, based on population estimates, may provide the most accurate prediction.

Figure 6: Panel A – life expectancy estimates for Australian men and women since 1890. Panel B – Population pyramid for Australia in 2016, demonstrating the proportion of population for each age group. Available from: https://www.aihw.gov.au/reports/life-expectancy-death/deaths-in-australia/contents/life-expectancy

 

If death from prostate cancer is compared with the likelihood of death from other conditions, the older a man, the greater is the likelihood that another condition will be the cause of his demise; in Australia in 2009, one in three male deaths was attributed to cardiovascular disease (28).

 

The following graphs (Figure 7) from the Australian Government website show approximately parallel increases for incidence and death from prostate cancer, estimated to be 23 years apart (Figure 7).  Consequently, if death is the endpoint being addressed, the patient’s life expectancy, based on his age and comorbidities, needs to be considered in the context of the natural history of his disease.

Figure 7: Estimated age-specific incidence (solid line) and mortality (dashed line) rates for prostate cancer, 2017 (Panel A), compared with 2007 (Panel B) (https://prostate-cancer.canceraustralia.gov.au/statistics)

TARGETING PROSTATE CANCER AT-RISK POPULATIONS

 

Major genetic epidemiologic studies published in the last two decades support the notion that prostate cancer may exist as clusters in families. In the 1980s, a Utah Mormon genealogy study found that prostate cancer exhibited the fourth strongest degree of familial clustering after lip, melanoma, and ovarian cancers (29). Prostate cancer, interestingly, had a higher familial association than either colon or breast carcinoma, to which patients are known to be predisposed by genetic or familial components.  A later study determined cancer pedigrees in 691 men with prostate cancer and 640 spouse controls and found that men with an affected father or brother were twice as likely to develop prostate cancer as men with no affected relatives (30). Although these findings strongly suggest that familial clustering of prostate cancer risk does exist, they did not address the underlying etiological mechanisms. Indeed, familial clustering can reflect either shared environmental and lifestyle risk factors, or a genetic mechanism, or both.

 

To determine what might distinguish hereditary prostate cancer from its sporadic counterparts, a number of clinical features of prostate cancer were examined by Carter, et al.(31). Clinical stage at presentation, pre-operative PSA, final pathologic stage, and prostate weight were examined in a series of approximately 650 patients divided among three categories. Individuals were classified as having hereditary disease if 3 or more relatives were affected in a single generation, prostate cancer occurred in each of 3 successive generations in either paternal or maternal lineages, or 2 relatives were affected under the age of 65 years. For the other groups, either no other family members were affected (sporadic disease), or other family members were affected but not to the extent found in families classified as hereditary.  In summary, no unique clinical or pathological characteristics distinguished hereditary prostate cancer in this group of patients.  This parallel between hereditary and sporadic prostate cancer also extends to the incidence of multifocality found in both of these categories.

 

These findings were supported by Brandt et al (2011) in an analysis of the nationwide Swedish Family-Cancer Database between 1961 and 2006. They found that the age-specific hazard ratio of prostate cancer diagnosis increased with the number of affected relatives and decreased with increasing age. The highest hazard ratios were observed for men <65 yr. of age with three affected brothers (approximately 23) and the lowest for men between 65 and 74 yr. of age with an affected father (HR: approximately 1.8). The hazard ratios increased with decreasing paternal or fraternal diagnostic age. The pattern of the risk of death from familial prostate cancer was similar to the incidence data (32). A similar study also from Sweden determined that a positive family history was a risk factor for developing prostate cancer and most pronounced in younger men (aged 45-49 years) (33). A vast array of molecular alterations implicated in sporadic and familial prostate cancer have been described (34)and reported to account for 30% of familial risk (35).

 

However, there are differences between hereditary and sporadic prostate cancers. The onset of hereditary prostate cancer is, on average, 6 years earlier than for sporadic cancer. Although the clinical course is in no way different and the pathological characteristics are the same in most instances (36), patients with a family history of germ-line mutations in the family-susceptibility genes BRCA1 and BRCA2 , in particular the latter, and G84E mutation in HOXB13(37), have a significantly increased susceptibility for developing this malignancy. Furthermore, these patients tend to present at a younger age, have more aggressive and disseminated disease with poorer survival outcomes [31-6](38-44).  Targeted screening of at risk men has been performed, with the IMPACT study reporting a higher positive predictive value of PSA and detection of intermediate- or high-risk disease in BRCA2 mutation carriers(45).

 

TESTS USED IN DIAGNOSING PROSTATE CANCER

 

In evaluating this issue, it is important to appreciate that the diagnostic approach is a two-step process that begins with the decision about whether or not to have a Prostate Specific Antigen (PSA) blood test (+/- other investigations) and, secondly, to confirm a suspected diagnosis of prostate cancer by biopsy for histopathology. Most men with a PSA level less than 10ng/ml will have a normal feeling prostate on digital rectal examination (DRE), hence the removal of DRE by non-urologists from many guidelines.

 

The FDA initially approved PSA testing in 1986 for monitoring the disease status of prostate cancer patients and, subsequently in 1994, it was endorsed as a screening method for prostate cancer (46). The PSA blood test is a continuous variable with no cut point (47)so that very low levels don’t completely exclude the possibility that prostate cancer is present(48-50), but the higher the serum PSA the greater the likelihood of prostate cancer being detectable. Importantly, PSA doesn’t distinguish between those who do and do not have cancer or identify those whose cancers will benefit from curative treatment. PSA increases with a number of conditions including prostate cancer, but the most common associated pathology is the non-cancerous condition benign prostatic hyperplasia (BPH) which is the cause, in most instances, of bladder outlet obstruction in men.

 

Factors Affecting PSA Measurements

 

Themedicationfinasteridewhich targets the 5-α-reductase type 2 enzyme and the more recently available drug, dutasteride, which inhibits both type 1 and type 2 enzymes, affect theconversion of testosterone to dihydrotestosterone (DHT) in prostatic cells. They reduce prostate volume with comparable effectiveness, with their designated clinical role being to decrease bladder outflow obstruction responsible for lower urinary tract symptoms (LUTS) present in a large number of older men. In reducing the benign prostatic hyperplasia (BPH) component of the prostate, both finasteride and dutasteride also reduce serum PSA levels by ~50% within 6 months of treatment.  However, with the influence of the non-cancer BPH component significantly reduced, PSA changes are more likely to indicate prostate cancer. For patients taking finasteride or dutasteride, an increase in PSA of >0.3 ng/ml from nadir is generally regarded as an indication for further investigation based on the findings of Marks et al (2006) who determined that applying this recommendation resulted in a 71% sensitivity and a 60% specificity for prostate cancer being detected in men receiving dutasteride (51). Use of dutasteride may also affect interpretation of multiparametric MRI (mpMRI) and require a reduced biopsy threshold(52).

 

Concerns with respect to finasteride use and subsequent prostate cancer were addressed by long-term data from the Prostate Cancer Prevention Trial. Results confirmed that finasteride reduced the risk of prostate cancer by about one third but also found that high-grade prostate cancer was more commonly found on biopsy in the finasteride group than in the placebo group. However, after 18 years of follow-up, there was no significant difference between-groups in the rates of overall survival or survival after the diagnosis of prostate cancer (53).

 

Other non-malignant causes affecting serum PSA levels include prostatic infection and ageing since prostates tend to become larger as men get older (54). Instrumentation of the prostate and urinary tract can also raise PSA levels (55)as can bacterial or severe prostatitis, both of these capable of resulting in sudden rises in this enzyme (Figure 8).

 

Testosterone supplementation is commonly used for hypogonadism and might intuitively complicate interpretation of serum PSA levels. However, available data suggests that testosterone supplementation does not significantly increase serum PSA (0.1 ng/mL; 95% CI -0.28 – 0.48)(56-57), prostate size, intraprostatic testosterone, or prostate cancer incidence and progression in men with pre-treatment serum testosterone higher than 5 – 7 nmol/L (144 - 202 ng/dl(58).  Testosterone therapy also causes minimal changes in lower urinary tract symptoms, with 77.5% of patients on supplementation having similar or improved symptoms for change in PSA of 0.44 (+/- 2.2)(59).

Figure 8: Factors Affecting Levels of Serum PSA(56-57) (60)

 

Efforts to improve the diagnostic accuracy of PSA have incorporated age-related reference ranges, which vary according to race (61-62)within and between countries. The normal age-related reference ranges are outlined below for European descent, Japanese, Chinese, Taiwanese, Singaporean and Korean men, as well as between Caucasian (Whites) and African-American (Blacks) men from the United States (Figures 9 – 14).

 

Furthermore, risk-stratification based on PSA for age is a newly adopted concept, recommended by the European Association of Urology and others, include longitudinal PSA testing for men with a PSA level >1 ng/ml at age 40 yr. or >2 ng/ml at age 60 yr. (63).

Figure 9: Age-based PSA Ranges for Men in Western Societies (19,61-62,64-66)

Figure 10: Age-based PSA Ranges for Japanese Men (67-70)

Figure 11: Age-based PSA Ranges for Chinese Men (67,71)

Figure 12: Age-based PSA Ranges for Taiwanese Men (67,72-74)

Figure 13: Age-based PSA Ranges for Singaporean Men (67,75-76)

Figure 14: Age-based PSA Ranges for Korean Men (67,77-78)

Attempts to improve the predictability of serum PSA for prostate cancer have included measuring the rate of PSA change (PSA velocity) and its relationship to the size of the prostate (PSA density) since prostates vary a lot in size and tend to become bigger as men age. This variable, but overall increase, in prostate size with ageing prompted the introduction of age-related PSAvalues by laboratories, based on the populations tested. The free or unbound PSA and its relationship to total PSA (free: total PSA) is another variation with the higher the free component, the lower the likelihood of cancer: most recently, the prostate health index (PHI) has become available and has been promoted. These are discussed in some detail (below).

 

Total PSA

 

Of the tests available, total serum PSA is generally regarded as having the greatest utility, maintaining its predictive value for the detection of prostate cancer (79)even after a first biopsy shows no evidence of cancer in which setting its performance characteristics are only slightly decreased (80). However, as stated above, PSA is far from a perfect test with most men with a serum PSA less than 10 ng/ml not having prostate cancer detected with biopsy, while conversely the possibility remains that prostate cancer may be present even with very low PSA levels.  In the Tyrol project, Pelzer et al (2005) found that prostate cancers detected in men with PSA levels <4 ng/ml were in younger patients and at lower stages (81).

 

In terms of reassurance, a PSA<1 ng/ml in a man aged 60 years has been reported to indicate an extremely low risk of clinically important prostate cancer in his lifetime. Although a 25-30 year risk of prostate cancer metastases could not be excluded by concentrations below the median at age 45-49 (0.68 µg/L) or 51-55 (0.85 µg/L), the 15-year risk remained low at 0.09% (0.03% to 0.23%) at age 45-49 and 0.28% (0.11% to 0.66%) at age 51-55 (82). This finding was supported by Aus et al who failed to find a single case of prostate cancer detected in 2950 screened men age 50-66 with a PSA <1ng/ml over a 3-year period (83).

 

Serum PSA Summary:

 

  • Is a continuous variable with no cut point (47)
  • Lodding et al (1998) found 15% of prostate cancers detected by investigating a PSA between 3 & 4 ng/ml had extraprostatic growth (49)
  • In the Tyrol project, prostate cancers detected in men with PSA levels <4 ng/ml were in younger patients and at lower stages with smaller prostate volumes (81)
  • Doesn’t indicate who will benefit from curative treatment (48)
  • Total PSA remains the single most significant, clinically-used predictive factor for identifying men at increased risk of harboring cancer (79)
  • For men 50-70 years, a PSA >1.5 ng/ml is a marker for greater than average risk up to 8 years (7.5-times greater risk versus 1.5 ng/ml or less) (79)
  • Sustained rises in PSA indicate a significantly greater risk of prostate cancer, particularly high-grade disease
  • A PSA <1 ng/ml in a man aged 60 years has been reported to indicate an extremely low risk of clinically important prostate cancer in his lifetime (50)
  • Not a single case of prostate cancer was detected in in 3 years in 2950 screened men with a PSA <1ng/ml (83)

 

PSA Velocity (PSAV)

 

PSA is a labile enzyme with falsely high readings as a result of ejaculation within the previous 48 hours, vigorous (non-sexual) exercise, urethral instrumentation, and prostatic infections, as well as different assays providing slightly different readings. Therefore, a single PSA level should not be relied upon to indicate an increase in level. A rate of change of PSA (PSAV) >0.75 ng/ml in year in the absence of another contributing cause equates with an increased risk of a patient having cancer (84).   Men taking the 5-α-reductase inhibitors, finasteride and dutasteride, have their serum PSA levels reduced by approximately 50% within 6 months. However, as stated above any sustained subsequent increase is more predictive for prostate cancer with an increase in PSA of 0.3 ng/ml from its nadir as a trigger for biopsy reported to provide 71% sensitivity & 60% specificity for prostate cancer for men who were receiving dutasteride (51).

 

For men not taking 5α reductase inhibitors, PSA increases >3.3% per annum have been reported to be associated with an increased risk of prostate cancer being detected by biopsy (19,65)and Makarov et al (2011) identified apreoperative PSA velocity >0.35 ng/ml/year to be associated with an increased risk of biochemical progression following radical prostatectomy (85).  A more sinister association was observed by D’Amico et al (2004) who found that a PSA increase >2 ng/ml in the year before diagnosis conferred a high risk of death from prostate cancer despite radical prostatectomy (86). Loeb et al (2012) confirmed the adverse significance of a rapidly rising PSA, reporting that patients with two PSA velocity measurements of >0.4 ng/mL/year had an 8-fold increased risk of prostate cancer and a 5.4-fold increased risk of Gleason 8-10 disease on biopsy, adjusting for age and PSA level (87). The same author also concluded from an analysis of the Baltimore Longitudinal Study of Ageing that, since PSAV rose continuously with increasing PSA and was significantly higher in cancers than controls for PSA levels <3 ng/mL and 3-10 ng/mL, the PSA level should be taken into account when interpreting PSAV (88).

 

PSA Velocity Summary:

 

  • A PSA increase >0.75 ng/ml per year increases the risk of prostate cancer (84); for men taking 5-α-reductase inhibitors  (finasteride & dutasteride) a PSA increase of 0.3 ng/mL per year increases the risk of prostate cancer
  • An increase in PSA of 0.3 ng/ml from nadir as a trigger for biopsy maintained 71% sensitivity & 60% specificity for prostate cancer in men receiving dutasteride (51)
  • A PSA increase of >3.3% per annum = an increased risk of cancer (19,65)
  • A preoperative PSA velocity >0.35 ng/ml/year = increased risk of biochemical progression following radical prostatectomy (85)
  • A PSA increase >2 ng/ml in the year before diagnosis = high risk of death from prostate cancer despite radical prostatectomy (86)
  • Men with two PSA velocity measurements of >0.4 ng/mL/year had an 8-fold increased risk of prostate cancer and 5.4-fold increased risk of Gleason 8-10 disease on biopsy, adjusting for age and PSA level (87)
  • An analysis of Baltimore Longitudinal Study of Aging concluded that, since PSA velocity rose continuously with increasing PSA and was significantly higher in cancers than controls for PSA levels <3 ng/mL and 3-10 ng/mL, the PSA level should be taken into account when interpreting PSAV (88)

 

 

Free/Total PSA

 

This test measures the percentage of free (or unbound) PSA in the blood and compares it with the percentage bound to proteins (α1 anti-chymotrypsin and α2 macroglobulin). Prostate cancer increases the amount of bound PSA.  The lower the ratio of free to total PSA or the percentage of free PSA, the higher the likelihood that the patient has prostate cancer. The proportion of free PSA in seminal fluid is much higher than in serum, consistent with its physiological role in liquefaction (89).  Although levels of bound-PSA do not significantly correlate with PSA in semen in young men, levels of free PSA do. With ageing, blood levels of complex-PSA, but not free-PSA, increase (90). The free/total PSA blood test can help to discriminate between patients with indeterminate PSA levels (4-10.0 ng/ml) indicating those who are at the greatest risk of having prostate cancer, in particular aggressive disease (91-92). However, as with all these modifications to PSA, the predictability remains less than perfect.

 

Free/Total PSA Summary:

 

  • Men with prostate cancer have a greater fraction of complexed PSA and a lower free PSA than men without prostate cancer
  • Free: Total PSA can be helpful in the case of a high PSA and a negative prostate biopsy
  • Free PSA is unstable: the assay must be frozen to -20°C within 3 hours otherwise the free fraction reduces
  • Chronic prostatitis may also cause a reduced Free: Total ratio

 

PSA Density

 

PSA density relates the concentration of serum PSA to the volume of the prostate and is thus a measure of serum PSA in relation to prostatic size (93). Most neoplastic prostate glands produce higher serum PSA levels per unit mass than do non-malignant glands. Consequently, a serum PSA of 5.0 ng/ml in a patient with a 20-gram prostate is more worrisome for cancer than that a PSA of 5.0 ng/ml in a man with a 60-gram prostate, especially if there is a predominance of transitional zone tissue (BPH) in the latter. To determine the PSA density, a PSA level is obtained and is divided by the volume of the prostate, as estimated by transrectal ultrasound (TRUS). Recent adoption of multiparametric (mp) MRI has allowed for determination of prostate volume as a standard reporting item. A value >0.15 ng/ml per gram of prostate tissue is considered worrisome for prostate cancer, and clinically used in nomograms to aid the urologist in estimating risk of prostate cancer. PSA density has been extended to include transition zone measurements in relation to the overall size of the prostate as the transition zone is the site in which BPH develops with ~25% of prostate cancers also arising in this zone. The larger the transition zone in relation to the overall size of the gland, the lower the likelihood of prostate cancer, other things being equal.

 

PSA Density Summary:

 

  • PSA Density = PSA divided by prostate volume determined by TRUS / mpMRI
  • The larger the transition zone, the lower the likelihood of prostate cancer
  • PSAD >0.15 ng/ml per gram is considered worrisome for prostate cancer
  • Problems with PSA density include:

(i) difficulty in defining the outline of the prostate accurately

(ii) variability in shapes not addressed by automated TRUS calculator estimations

 

Prostate Health Index

 

A further variation on the PSA blood test is the Prostate Health Index or phi, formulated by having the value of a truncated form of the PSA molecule (proPSA, greater production by most cancers than benign tissue) as the numerator and the free PSA value as the dominator multiplied by the total PSA level to give a phireading. phiis claimed to better predict prostate cancer risk than the total PSA. A phi-based nomogram in an external validation study performed with 75.2% accuracy (94). Furthermore, phi has been reported to aid in predicting pT3 disease (2.3%) and/or pathologic Gleason score ≥ 7 (2.4%) although decision curve analyses deduced these were not of greater clinical net benefit (95). A potential advantage of phiis that it stratifies according to risk. However, health economic analyses to determine clinical benefits of phi are yet to be realized(96).

 

Prostate health index [phi] = [−2]proPSA / fPSA) × PSA1/2

 

  • For PSA 2–10 ng/ml, sensitivity, specificity and AUC (0.703) of phiexceeded those of total PSA and % fPSA. Increasing phiwas associated with an increased risk of prostate cancer (97). These estimates have been confirmed in multiple studies (AUC 0.67-0.81 c.f. PCA3 0.73, %fPSA 0.60-0.65, tPSA 0.50-0.52)(94,98-99), resulting in a consistent estimate of approximately 20-30% of avoided biopsies if phi is used instead of %fPSA(100-101). A meta-analysis estimated prostate cancer detection with sensitivity of 90% and specificity of 31.6%, and was better overall than PSA and %fPSA for PSA 2 – 10ng/ml(102).
  • Including the prostate health index in a multivariable logistic regression model based on patient age, prostate volume, digital rectal examination and biopsy history significantly increased predictive accuracy by 7% from 0.73 to 0.80 (p <0.001) (103).
  • phi0-22.9            =          low probability of prostate cancer      (8.4%)

23-44.9            =          moderate probability of cancer           (21%)

>45                 =          high probability of cancer                   (44%)

  • phi-density (PHID), calculated similarly to PSA density, may also improve diagnostic accuracy of clinically-significant prostate cancer (AUC 0.82) compared to phi (0.79), %fPSA (0.79) and PSA (0.70)(104).

 

Four-kallikrein (4K) Panel

 

Another recent variation on the PSA blood test is the four-kallikrein (4K) panel, determined by a combination of kallikrein-related peptidase 2 (hK2), intact PSA, and free and total PSA as well as with clinical data (age, DRE findings, previous biopsy results). The 4K score has been shown to predict biopsy outcome to avoid unnecessary biopsies as well as predict distant metastasis at 10 years.

 

  • Among European men with serum PSA 3 – 15 ng/ml, 4K score showed similar diagnostic performance (AUC 0.69 any prostate cancer, 0.72 high-grade prostate cancer) to phi (AUC 0.70 any prostate cancer, 0.71 high-grade prostate cancer) and potentially saved 29% of performed biopsies(105). These findings have been confirmed in multiple studies, including a Swedish community cohort(106).
  • When applied to a USA cohort, 4K score performed better (AUC 0.82) than PCPT clinical risk calculator (AUC 0.74), equating to a potential 30-58% reduction in biopsies for delayed diagnosis of 1.3-4.7% of Gleason≥7tumors (107). Furthermore, accuracy was maintained between African American and Caucasian groups.
  • The 4K score was applied to patients enlisted in the ProtecT study and showed superior diagnostic accuracy compared with PSA for any cancer (AUC 0.719 vs. 0.634) and high-grade cancer (0.82 vs. 0.74) and potentially reduced unnecessary biopsies by 42%(108).

 

Summary: Prostate Specific Antigen (PSA) & Derivatives

 

  • Is a continuous variable with no cut point (47)
  • Doesn’t distinguish between those with and without cancer or identify those with cancer who will benefit from curative treatment (48)
  • PSA Velocity = rate of change of PSA: A PSA increase >0.75 ng/ml in year = an increased risk of having cancer  (84)
  • PDA Density: PSA density = PSA divided by prostate volume determined by TRUS
  • Free: total PSA: The higher the free component and the ratio of free to total PSA, the lower the likelihood of cancer but chronic prostatitis may also cause a reduced Free: Total ratio
  • Prostate Health Index (phi):may predict the risk of prostate cancer better compared with total PSA, but its role in prostate cancer screening is not defined.
  • Total PSA = the single most significant, clinically used predictive factor for identifying men at increased risk of harboring cancer (79)

 

Digital Rectal Examination

 

Traditionally, palpation of the prostate by digital rectal examination (DRE) was the manner by which a diagnosis of prostate cancer was suspected. In historical series, up to 50% of palpable masses were attributable to prostate cancer (17,109-110). Although DRE by itself is a poor method for diagnosing this malignancy (111-112), especially when performed by non-urologists, it does still have an important diagnostic role, hence its variable inclusion in prostate cancer guidelines (recommended only for urologists mostly), as up to 25% of tumors are detected in men with normal PSA levels (113). Unfortunately, when a prostate cancer is diagnosed based on a palpable tumor, the risk of the patient already harboring metastatic or locally advanced malignancy is considerable(114-116).  However, a PSA-based prostate cancer detection strategy which omits DRE runs the low risk of missing some curable cancers (49).

 

The PCA3 Test

 

The non-coding RNA PCA3, originally called DD3, is highly specific to prostate cancer, with over-expression(117-120)in a number of different cohorts.  The first part of a voided urine specimen is collected immediately following firm rectal examination or prostatic massage (121-122)and PCA3 RNA measured using a PCR-based assay. One criticism of the PCA3 test is that is unlikely to obtain prostatic fluid from the anterior part of the prostate, mirroring a deficiency with TRUS-guided biopsies obtained via the rectum, which are also posteriorly-focused, especially in large prostate glands.  Although the “PCA3 urine test” has been reported to improve identification of serious disease compared with total PSA in a pre-screened population (Table 2), its role in initial assessment of patients suspected of having prostate cancer has yet to be established (123-124). A prospective multicenter validation trial to assess the diagnostic performance of PCA3 determined a positive predictive value of 80% for initial biopsy, and negative predictive value at repeat biopsy of 88%, while the addition of PCA3 to available risk calculators improved risk prediction of overall and high-grade cancer(125).

 

Table 2: PCA3 Results in Post-Prostatic Massage Urines (118,120,125-129)

Study Sensitivity Specificity Neg Predictive Value Number
Hessels et al, 2003 67% 83% 90% 108
Fradet et al, 2004 66% 74% 84% 517
Tinzl et al, 2004 82% 76% 87% 158
Van Gils et al, 2007 65% 66% 80% 534
Van Gils, et al 2007 65% 82% 80% 67
Salami et al, 2013 93% 37% 92% 45
Wei, et al 2014 (initial biopsy, PCA3 > 60) 42% 91% PPV 80% 562
Wei et al 2014 (repeat biopsy, PCA3 <20) 76% 52% 88% 297

 

Attempts to analyze PCA3 and other biomarkers in prostatic fluids, such as semen(130-131), have shown comparable diagnostic accuracy (132)but patient recruitment and clinician acceptance is challenging.

 

Recently, data from analysis of the fusion gene TMPRSS2:ERG and PCA3 from prostatic fluid obtained following firm digital rectal examination/prostatic massage, has been combined with serum PSA to produce a test which is being marketed commercially. Published supportive data is limited but preliminary findings indicate that the combination provides an 80% sensitivity and 90% specificity with an AUC of 0.88 for the 3 parameters(129,133-134).

 

However, Stephan et al.(135)examined PCA3, TMPRSS2:ERGand phiin an artificial neural network. The addition of TMPRSS2:ERG to PCA3 in urine following firm digital rectal examination only marginally improved detection of prostate cancer in110 men compared with 136 with non-cancer. PCA3 had the largest AUC (0.74) which was not significantly different to the AUC of phi(0.68) although the latter showed somewhat lower specificities than PCA3 at 90% sensitivity.  A combination of PCA3 and phionly moderately enhanced diagnostic power with modest AUC gains of 0.01-0.04 for prostate cancer at first or repeat prostate biopsies. These findings were not reproduced by Salami and colleagues in the USA, where PCA3 demonstrated high sensitivity (93%, AUC 0.65), while TMPRSS2:ERG had higher specificity (87%, AUC 0.77) compared to serum PSA (AUC 0.72). A multivariate algorithm optimized cancer prediction (AUC = 0.88; specificity = 90% at 80% sensitivity) (129). In a prospective multicenter study of PCA3 and TMPRSS2:ERG prior to biopsy, Sanda and colleagues reported a 33-39% specificity at 93% sensitivity, which was predicted to reduce 42% of unnecessary biopsies and conferred a cost benefit for younger men(136).

 

It is likely that future clinical practice will integrate molecular markers into predictive calculators, such as the Prostate Cancer Prevention Trial (PCPT) or ERSPC calculators, to improve diagnostic accuracy above crude traditional markers such as family history or clinical examination findings (137).

 

Magnetic Resonance Imaging (MRI)

 

 

MRI use in prostate cancer is rapidly evolving. Potential applications and benefits include are summarized in Table 3.

 

Table 3. Utility of MRI in Prostate Cancer(138-141)

Scenario Potential Benefits
1) Triage prior to biopsy ·       Avoid unnecessary biopsy

·       Provide target(s) for biopsy

·       Assessment of anterior and apical areas that are poorly sampled by TRUS biopsies obtained via the rectum

·       Increase detection of clinically significant cancer

·       Minimize detection of insignificant cancer

2) Patients with prior negative biopsy ·       Provide target for biopsy

·       Assessment of anterior and apical areas that are areas poorly sampled by TRUS

·       Increase detection of clinically significant cancer

·       Minimize detection of insignificant cancer

·       Decrease unnecessary repeat biopsy

 

As an initial form of detection, MRI has the potential to improve the sensitivity of detection of intermediate and high-risk prostate cancer, especially in the anterior zone of the prostate, where cancers may not be sampled using transrectal ultrasound guided biopsy techniques. However, in some countries cost is still a handicap to widespread application.  Interpretation of prostate imaging with different sequences (summarized in Table 4), using a “multiparametric” approach, requires expertise and collaboration, most commonly according to a structured reporting scheme, prostate imaging-reporting & data system (PI-RADS), which has since been updated to PI-RADS v2  (142-144). A PI-RADS score is assigned to each individual lesion using T2 weighting, diffusion-weighted imaging (DWI), and dynamic contrast enhancement to assign scores based on a Likert (5-point) scale based on the probability of clinically significant malignancy: PI-RADS 1 very low; PI-RADS 2 low; PI-RADS 3 intermediate; PI-RADS 4; PI-RADS 5 very high (145). A summary of the standardized anatomical description map and PI-RADS scoring specifications are shown Figures 15 and 16).  For lesions in the peripheral zone DWI is the dominant sequence, and for the transition zone T2 dominant.

 

Table 4: MRI Imaging Sequences

Use
T1-weighted Detection of hemorrhage post biopsy as hyperintense

Detection of bone metastasis

Detection of abnormal lymph nodes

T2- weighted Demonstrates zonal anatomy.

Sensitive but non-specific as prostate cancer, prostatitis, atrophy, BPH, and changes after treatment (e.g., radiation induced arteritis) are hypointense

Dominant sequence for PIRADS scoring of transitional zone.

Diffusion-weighted imaging (DWI) Prostate cancer demonstrates restricted diffusion, appearing hyperintense at high b values and hypointense on ADC map

Dominant sequence for PIRADS scoring of peripheral zone

Dynamic contrast enhancement (DCE) Prostate cancer shows early enhancement and early washout
Spectroscopy Requires extra time for acquisition and may not add diagnostic value.

Not frequently used.

Citrate is reduced, whereas choline is increased in prostate cancer

Figure 15: Prostate Map for Description of Lesions Detected on mpMRI According to PIRADS Classification

Figure 16: Prostate Map for Description of Lesions Detected on mpMRI According to
PIRADS Classification for Peripheral (right) and Transition (left) Zone Lesions.

CLINICAL USES OF MRI IN PROSTATE CANCER

 

Triage Prior To Biopsy

 

The diagnostic accuracy of MRI for the detection of prostate cancer varies widely across studies, with sensitivities from 58-96% and specificity from 23-87% (140-141,146-148). Such variability can be explained by different equipment, level of experience, lack of standardization in the early series, different reference standards, different sequencing protocols and definitions of clinically significant cancer. It is evident that accuracy has been improving in the more recent series, and good standardization of reporting has been achieved with the use of PIRADS V2. A meta-analysis of 21 studies including 3857 patients using PIRADS V2 showed a sensitivity of 89% and specificity of 73% (149). Of note, some studies only considered clinically significant prostate cancer while other considered any prostate cancer (149).

 

The PROMIS trial of 576 men assessed the capacity of mpMRI to identify men with clinically significant prostate cancer prior to prostate biopsy and compared the diagnostic accuracy of mpMRI to a 10-12-core systematic TRUS biopsy using a template transperineal prostate biopsy with cores taken every 5mm as the reference standard (148). mpMRI was significantly more sensitive than TRUS biopsy via the rectum in all 3 definitions of significant cancer used (Table 5). As a triage test mpMRI performed prior to biopsy and reserving it to patients with suspicious findings, mpMRI would have avoided biopsying 27% of patients at a risk of missing 7-12% of significant cancers - depending on the definition used.

 

Table 5. PROMIS Trial Results (148)

Definition mpMRI TRUS 10-12-core
Gleason ≥4+3 or cancer ≥6mm Sens: 93%

Spec: 41%

Sens: 48%

Spec: 96%

Gleason ≥3+4 or cancer ≥4mm Sens: 87%

Spec: 47%

Sens: 60%

Spec: 98%

Gleason ≥3+4 Sens: 88%

Spec: 45%

Sens: 48%

Spec: 99%

 

The PRECISION study was a multi-centre pragmatic trial that randomized 500 patients with elevated PSA and/or abnormal DRE to TRUS prostate biopsy (10-12 cores) or a mpMRI. Patients in the mpMRI group underwent a targeted biopsy only if lesion(s) PIRADS ≥ 3 were identified. The targeted biopsy could be performed with software or cognitive fusion and could be transrectal or transperineal. The detection of clinically significant prostate cancer (defined as Gleason 3+4 or greater) was 26% in the standard biopsy group and 38% in the mpMRI group. This increased detection occurred despite the fact that 28% of patients in the mpMRI group were not biopsied because the study was reported as PIRADS 1-2 but still included in the population/denominator. Conversely, the detection of Gleason 3+3 cancer was 22% in the standard biopsy and 9% in the mpMRI group. Clinically significant cancer was detected in 12% of PIRADS 3, 60% of PIRADS 4 and 85% of PIRADS 5 lesions, similar to that seen in previous studies. Of note, the centers contributing the majority of patients had significant prior experience with prostate mpMRI reporting and targeted prostate biopsies. Whether centers with limited experience can achieve similar results remains to be demonstrated.

 

Patients with Prior Negative Biopsy

 

MRI has shown been shown to be useful in the subset of patients with prior negative biopsies. For these patients, the biopsy detection rate is approximately 30%, decreasing with each subsequent biopsy procedure (150-151).MRI followed by MRI-guided biopsies identifies prostate cancer in 41-59% (152-153). A recent review showed that across 16 studies MRI guidance improved the absolute detection of clinically significant prostate cancer between 6-18% in patients with previous negative TRUS biopsy (154).

 

DEFINITIVE DIAGNOSIS REQUIRES BIOPSIES

 

Prostate biopsy is required for the definitive diagnosis of prostate cancer. Systematic TRUS-guided biopsies have been the standard for the past decades but concerns about missing significant cancer and the risk of sepsis are changing the landscape. While TRUS imaging permits spatial positioning for systematic sampling, by itself it has low accuracy in detecting suspicious areas.

 

The number of biopsy cores taken is important with the chance of missing a cancer by standard sextant biopsy estimated to be approximately 25% (155)so that, more recently, the numbers of cores recommended are at least 10-12.  In addition, it is advocated that biopsies should be directed laterally and that they should include the anterior horns of the peripheral zone (155-158). Still, recent studies have shown that systematic TRUS biopsies performed via the rectum miss approximately 50% of clinically significant cancers (139,148,159). The introduction of mpMRI is changing this situation at least in terms of missing significant cancer.

 

One of the problems facing clinicians has been when to stop from recommending biopsying not only in terms of patient age and overall life expectancy but also with respect to the increasing likelihood of a positive histological diagnosis in those biopsied.  Indeed, a continually increasing probability of death from prostate cancer was observed among men of all ages with a PSA of 3.0 ng/ml in Baltimore Longitudinal Study of Ageing (160)of 849 men, 122 with and 727 without biopsy-confirmed prostate cancer. However, no participants between 75 and 80 years old with a PSA lower than 3.0 ng/ml died of prostate cancer. And not unexpectedly, the time to death or diagnosis of aggressive prostate cancer after age 75 years was not significantly different between PSA categories of 3 to 3.9 and 4 to 9.9 ng/ml. Of the 108 subjects older than 75 years with a PSA of 3 ng/ml or greater, 10 died of prostate cancer and 18 had high risk disease. In this group, 90 men did not have a diagnosis of high risk prostate cancer, including 75 who were never diagnosed with cancer (median time to censoring 12.5 years) and 15 who were diagnosed with non-high risk cancer (median time to censoring 17 years) (160). Therefore, many guidelines recommend against PSA testing among men older than 70 with a life expectancy of less than 7 to 10 years.

 

Routine practice for biopsies taken via the rectum involves peri-operative antibiotic prophylaxis. TRUS biopsies can be performed under local anesthesia or sedation. Rectal cleansing with povidone-iodine is recommended to decrease the risk of sepsis (161).

 

Changing Morbidity of Biopsy Diagnosis

 

Periprocedural symptoms such as hematuria, rectal bleeding and hematospermia are frequent, being experienced by over 50% of men having TRUS biopsies performed via the rectum but are almost always benign and self-limiting (162-164). Infectious complications following this procedure are less common but are being reported more often, with the causative mechanism believed to be inoculation of the prostate, blood vessels and urine with bacterial flora from the rectal mucosa and subsequent systemic dissemination (162,165-166).   There has been concern expressed that hospital admissions due to post-TRUS biopsy may be rising, with one study reporting a 3-fold increase from 0.55% across 2002-2009 to 2.15% across 2010-2011 (162,167-168). Changing bacterial resistance patterns and antibacterial practices have contributed to the spectrum of infectious complications with the infection rate being much higher in certain population groups such as men who have been taking antibacterial drugs prior to the biopsy and people who have been in South East Asia and Mediterranean countries within the past 6-12 months (168-169). Not surprisingly there is wide variation in the reported incidence of overall infectious complications from 0.1% to 7% and of sepsis from 0.3% to 3.1% across studies (166,170).

 

A prospective New Zealand study reported that drug resistance rates for patients who required intensive care admission for sepsis following TRUS biopsy were 43% for gentamicin, 60% for trimethoprim-sulphamethoxazole (60%) and 62% for ciprofloxacin as well as 19% for all 3 agents in combination.  E. coli sequence type 131 clone was implicated as being particularly problematic, accounting for 41% of all E. coli isolates after TRUS biopsy (171).  Fluoroquinolone resistance in rectal cultures has been reported to predict infectious complications following TRUS biopsy (172). The changing patterns of drug sensitivities and reports of low resistant rates to drugs such as carbapenems for patients with unresolving sepsis (173)has resulted in some advocating for the use of these drugs as prophylactic agents just prior to TRUS biopsy (174-175). However, adoption of such a strategy runs the risk of decreasing the number and effectiveness of those pharmaceutical agents currently kept in reserve for patients with overwhelming sepsis (175).

 

The transperineal approach has emerged as an alternative with significantly decreased risk of infections complications, albeit requiring specialized equipment, general anesthesia in most centers, increased operative time, an increased risk of urinary retention and potential nerve damage affecting erectile ability. A notable advantage of the transperineal approach is better sampling of the anterior zone of the prostate (174).

 

MRI also allows one to perform targeted biopsies, thereby increasing the detection of significant cancer. The main types of guided prostate biopsy techniques following diagnostic imaging with MRI include cognitive fusion (visual estimation from clinician’s interpretation of TRUS and mpMRI images), MRI-guided (biopsy performed under MRI guidance), fusion software (software integrating MR images on to the TRUS screen to guide biopsy needle to target index lesions), and robotic (automatic fusion and alignment for clinician). A particular issue with biopsying performed under real-time MR imaging is cost because this approach uses MR equipment which otherwise would be used for other purposes. These options are summarized in Table 6.

 

Table 6: Approaches for MRI-guided Targeted Prostate Biopsy

Description Characteristics References
Cognitive fusion (visual estimation) Manually directed based on MRI

TRUS or TP

Low cost

Operator dependent

Sciarra 2010

Lee 2012

Panebianco 2015

In-gantry
(real time) MRI-guided biopsy
MRI-compatible biopsy gun used and trajectory established. Biopsy gun fired and sampling confirmed

TRUS or TP

High cost with each procedure

Steep learning curve

Highest precision

 

Overduin 2013

Penzkofer 2015

Schimmöller 2016

Yaxley 2017

MRI-TRUS software fusion biopsy Software assisted targeting of lesions

TRUS or TP

Initial cost outlay

Ongoing costs similar

Good accuracy

Porpiglia 2016

Siddiqui 2015

Meng 2016

Robotic-Assisted Potentially less operator dependent

TRUS or TP

Initial cost outlay Tilak 2015

 

In a meta-analysis of 11 studies Wegelin et al. compared the prostate cancer detection rates of cognitive-fusion, in-gantry, and TRUS software-fusion biopsy. In-gantry biopsy had a higher overall detection rate than cognitive-fusion, but the detection of clinically significant cancer was not different across the 3 techniques. Yaxley et al. performed a retrospective review comparing in-gantry MRI-guided biopsy to cognitive TRUS biopsy. In 595 PI-RADS 3-5 lesions, there was a high prostate cancer detection rate with no difference across biopsy methods (176). While the advantages of obtaining an MRI prior to biopsy are clear, up to 13% of clinically significant tumors can be missed when only targeted biopsies are performed (177-178). Moreover, this figure may be higher for lower volume centers with limited experience in MRI interpretation and MRI-guided biopsies. Consequently, many practitioner’s biopsy both index lesions seen with MRI as well as systematically sampling all parts of the prostate with 12 or more biopsies. In doing so, cognitive or in-gantry approaches are used for index lesions with a template employed to ensure correct special placing of biopsy needles for systematic sampling of the whole of the prostate.

 

Histopathological Assessment

 

The biopsy result provides important information for the patient and clinician on which to base management decisions (179).  Important prognostic information on biopsy assessment include tumor quantification values (fraction of positive cores i.e. the number of positive cores versus the number of cores submitted and the percentage or length in mm of cancer in intact positive cores), cancer grade (Gleason score in each positive core and ISUP [International Society of Urological Pathology] grade) and presence or absence of perineural invasion, lymphovascular invasion, intraductal carcinoma, and extraprostatic extension (180). Increasing tumor burden and poor histological differentiation are associated with a higher risk of metastatic disease, an increased chance of post-treatment failure, and a worse overall prognosis (181-183).

 

Histological analysis is the ‘gold standard’ for classifying prostatic adenocarcinoma. Using architectural patterns, the tumor is assigned a Gleason score and ISUP grade between 1 and 5, with higher numbers representing less differentiated, more aggressive tumors (see Table 7). A single prostate can harbor multiple foci of different histologic patterns of adenocarcinoma, and it is possible to have Gleason grade 3, 4 and 5 patterns in the same specimen: 85% of prostate tumors are multifocal. The Gleason score and ISUP grade are generated by combining the values of the first and second most common (dominant and subdominant) grades assessed by the uropathologist using light microscopy. In needle biopsies, the Gleason score and ISUP grade are calculated using the most common and highest grade of cancer (184). These values provide s important prognostic information.

 

Table 7. The International Society of Urological Pathology (ISUP) Grading System (185)

ISUP grade Gleason scores Definition
Grade 1 2-6 Only individual discrete well-formed glands

 

Grade 2 3+4=7 Predominantly well-formed glands with

lesser component of poorly formed/ fused/ cribriform glands

Grade 3 4+3=7 Predominantly poorly formed/fused/ cribriform glands with lesser component of well-formed glands

 

Grade 4 4+4=8 Only poorly formed/fused/cribriform glands
3+5=8 Predominantly well-formed glands and

lesser component lacking glands (or with necrosis)

5+3=8 Predominantly lacking glands (or with

necrosis) and lesser component of

well-formed glands

Grade 5 9-10 Lacking gland formation (or with necrosis)

with or without poorly formed/fused/

cribriform glands

 

The presence of Gleason grade 4 or greater histology carries a significantly poorer prognosis (186-187). It has been shown that Gleason score 4+3 tumors behave much worse than Gleason score 3+4 tumors and that there is a biological continuum within Gleason score 7 tumors with the proportion of pattern 4 cancer that is reflected in clinical outcome (188).

 

In the large majority of instances, gray-scale TRUS does not permit differentiation between cancer and non-cancer so TRUS and transperineal biopsies are taken blindly. Consequently, there is a possibility that small tumors may be missed, despite careful spatial positioning of biopsy needles with multiple cores taken. Furthermore, in large glands especially, the anterior part of the prostate may be poorly sampled via the transrectal route so, for these reasons, it is not surprising that the histology from biopsies and radical prostatectomies may differ. In these instances, the Gleason score from the radical prostatectomy specimen is usually higher (upgrading) but downgrading is also observed.

 

Recently, the International Society of Urological Pathology (ISUP) proposed a new Grading system in order to improve prognostication of tumor grade, as well as improve patient education (184,189-191). Although the term “grade groups”has been used for these prognostic categories, it has been shown to be erroneous as they are not groupings of grades but groupings of scores (184,189). Furthermore, these categories were the result of a consensus conference organized by the ISUP for the purpose updating the ISUP modified Gleason scoring system of 2005 and as such, the new ISUP grades are based on the 2005 ISUP modified Gleason scores (Table 7).

 

These grades have been validated in surgical cohorts and show distinct patterns of recurrence free progression (RFP) depending on the highest Grade within the RP and biopsy histology (190-191).

 

PROSTATIC INTRAEPITHELIAL NEOPLASIA [PIN]

 

Prostatic intraepithelial neoplasia [PIN] is believed to be a precursor of prostate cancer, given the strong association between high grade PIN and prostatic adenocarcinoma (192-194). The presence of high grade PIN is often indicative of the presence of prostate cancer. It has been shown that more than 80 percent of prostates with adenocarcinoma also contain high-grade PIN (PIN-11 & III). High-grade PIN has cytologic features resembling cancer and carries many of the genetic alterations of prostate cancer. The finding of high-grade PIN alone in a biopsy has been cited as an indication to proceed with repeat biopsies given the high co-frequency between high-grade PIN and carcinoma. However, in current practice, the predictive value of PIN in finding cancer on subsequent biopsies has declined, probably due to the extended biopsy techniques yielding higher rates of initial cancer detection (195). A diagnosis of PIN by itself is certainly insufficient for a patient to undergo either radical prostatectomy or radiotherapy.

 

ATYPICAL PROSTATIC GLANDULAR PROLIFERATIONS

 

Foci of atypical glands, also labeled ‘atypical small acinar proliferation of uncertain significance’, have features suspicious for, but not diagnostic of, cancer. These encompass a variety of lesions including benign mimickers of cancer, HGPIN, and small foci of carcinoma which, for a variety of reasons, cannot be accurately diagnosed. The reported incidence of these lesions on prostate needle biopsies is 1.5% to 5.3% (195). Patients with atypical glands on needle biopsy have a high risk of harboring cancer. The reported incidence of prostate cancer from repeat biopsies has ranged from 34 to 60% (196). Following an atypical diagnosis, biopsies need to be repeated (197).

 

TNM STAGING SYSTEM

 

Once a diagnosis of prostate cancer is made, it must be determined whether the patient is a candidate for potentially curative treatment (surgery or radiation). This depends upon several factors, including general health and projected longevity in conjunction with the likelihood that the cancer is still localized within the prostate and has not yet metastasized. The most important factor, however, is the patient’s decision after he has considered the ‘pros and cons’ of the various choices as they relate to him (see below).

 

Currently, the TNM system is used for staging (Table 8), and prostate cancers can be assigned both a clinical stageand, subsequently should the prostate be removed surgically, a pathologic stage. This differentiation is important with the clinical and pathological stage designated by the letters ‘c’ and ‘p’, respectively, preceding the stage denotation (e.g. cT2a = clinically, tumor is palpably involving one lobe of the prostate or less).

 

Table 8: TNM Staging Classifications [per American Joint Committee on Cancer (AJCC) 8th Edition 2016)(198)

Primary Tumor
     Tx

T0

Primary tumor cannot be assessed

No evidence of primary tumor

     T1 Clinically inapparent tumor not palpable not visible by imaging
     T1a Incidental tumor in < 5% of TUR tissue
     T1b Incidental tumor in > 5% of TUR tissue
     T1c Needle biopsy prompted by elevated PSA
     T2 Organ confined
     T3 Tumor extends beyond the prostatic capsule
     T3a Extracapsular, unilateral and bilateral or microscopic invasion of bladder neck
     T3b Tumor invades seminal vesicles (s)
     T4 Tumor invades external sphincter, rectum, pelvic side wall
Lymph Nodes
     Nx

N0

Regional nodes were not assessed

No regional (below level of bifurcation of common iliac arteries) nodes

     N1 Regional node metastases – including pelvic, hypogastric, obturator, iliac, sacral
Distant Metastases
    Mx

M0

Regional nodes not assessed

No Metastases

    M1

M1a

M1b

M1c

No distant

Non-regional lymph nodes (outside true pelvis)

Bone(s)

Other site(s) with or without bone disease

 

POTENTIAL BENEFITS & HARMS FROM PSA TESTING

 

One of the most contentious topics in medicine is whether or not to test for prostate cancer. The key question that needs to be answered is whether a diagnosis of prostate cancer is going to benefit the patient with the qualification that the diagnostic process and treatment should not be worse than the unwanted effects of the disease. Determining who will benefit from testing is very difficult as it is impossible to know exactly how long an individual patient will live and generally both patients and clinicians tend to be optimistic in their estimations.

 

Early Diagnosis and Treatment With Curative Intent And Prevention Of Subsequent Death From Prostate Cancer

 

In addition to attributing a slow but continuing reduction in prostate cancer mortality in many Western countries to, at least in part, widespread PSA testing, most of the evidence proffered in support is from low-level cohort studies, many of which have been retrospective. One notable, largestudy undertaken prospectively has been in the Tyrol. Unlike in the rest of Austria, PSA testing has been freely available in Tyrol since 1993 for men 45-75 years with 86.6% of eligible men having been tested at least once since its inception (199). Compared with the rest of the country, there has been a decreasing trend in prostate cancer mortality which, in 2005, was significantly greater in the Tyrol compared with the rest of Austria (P = 0.001). Prostate cancer deaths were 54% lower than expected in this region compared with the rest of Austria, with a significant migration to lower stage disease. These better results in Tyrol have been attributed to early detection, consequent down-staging and effective treatment.

 

However, the evidence for and against PSA screening is usually based on the findings from 6 mass or whole of population screening trials and meta-analyses of their findings. These studies were the Prostate Lung, Colorectal and Ovarian (PLCO) Screening Trial (200-201), the European Randomized Study of Screening for Prostate Cancer (ERSPC)(48)  (202), Göteborg (203), Norrköping (204), Stockholm (205)and Quebec trials (206).

 

The studies were very different in design and in adherence to protocols. For example, men were invited only once in Stockholm Study and a minority of those with screen-detected prostate cancer were treated with curative intent (205).  The participation rate was only 24% in the Quebec study (206). The Norrkoping Study commenced in 1987 with DRE as the only screening test performed up to the third (1993) and the final fourth screening time (1996) when PSA was included. Fewer than 500 men had two PSA measurements & none had more than two. Furthermore, final results were adjusted for the large difference in age at randomization between the study groups (204).

 

Thus, in terms of trials with reasonable rigor, there are only 3 viz. the ERSPC, the Göteborg (which is also included as part of the larger ERSPC study) and the PLCO trial (Table 9). In the PLCO trial only 85% in the screening arm had a PSA test. In addition, more than 80% in the control arm reported having a PSA test, significantly contaminating this arm (200,207). Furthermore, the follow-up for these trials varied greatly with only one (Göteborg) having an adequate median follow-up period, detailed below.

 

PLCO:             median 11.5 years,   maximum 13 years (201)

ERSPC:          median 9.8 years,     maximum 11 years (202)

Göteborg:      median 14 years,      maximum 14 years (203)

Norrköping:     median 6.3 years,       maximum 20 years (204)

Stockholm:      median 12.9 years,     maximum 15 years (205)

Quebec:          median 7.9 years,       maximum 13 years (206)

 

Table 7: Comparison of ERSPC, PLCO and Göteborg Trials

ERSPC PLCO Göteborg
Number studied 162 243 76,693 20,000
Recruitment sites 8 countries 10 US centers one
Age 50-69 55-74 50-64
PSA screening interval 4 yearly yearly x6 DRE x4 2 yearly
Biopsy trigger 3.0 ng/ml >4 ng/ml 3.4, 2.9, 2.5 ng/ml
Contamination rate

(PSA testing in control group)

15% 52% 3%

 

Since the studies are so different in so many ways, the validity of including them in a meta-analysis has been questioned (208). Given the long natural history of prostate cancer in comparison with those of other malignancies and the prevalence of the diseasewith increasing age, few would advocate screening each and every member of a population (209-211)i.e. mass population screening as reported in these trials

 

SUMMARY OF MORTALITY FINDINGS FROM THE THREE MOST RELEVANT STUDIES

 

  • None of these trials had adequate statistical power to detect an overall survival benefit with PSA screening
  • Deaths from conditions other than prostate cancer dominated causes of death undermining ability to show an advantage for PSA screening

 

  • PLCO- At a median follow-up of 11.5 years, of 76 685 men randomized (38,340 in the intervention arm and 38,345 in the control arm) (201). Approximately 92% of the study participants were followed for 10 years and 57% for 13 years.

-   deaths from all causes other than prostate, lung, and colorectal cancers were 5783/38,340 (15%) in the intervention arm: 5982/38 345 (15.6%) in the control arm

-   of those who died, 158/5783 (2.7%) & 145/5982 (2.4%) in the control arm, died from prostate cancer, respectively

-   cumulative mortality rates from prostate cancer in the intervention and control arms were 3.7 and 3.4 deaths per 10 000 person-years

 

  • ERSPC- At a median follow-up of 11 years, 31,318 of 162,388 (19.3%) of men between 55 & 69 yr. who underwent randomization had died [154] (202)

-   13,917/72,891 (19%) in screening group: 17,256/89,352 (19%) in control group

-   of those who died, 299/13,917 (0.4%) & 462/17,256 (0.5%) died from prostate cancer, respectively

-   the absolute reduction in mortality in the screening group was 0.10 deaths per 1000 person-years or 1.07 deaths per 1000 men who underwent randomization.

-   to prevent one death from prostate cancer at 13 years of follow-up, 781 men would need to be invited for screening and 27 cancers would need to be detected (212)

 

  • Göteborg- At a median follow-up of 14 years, 3,963 of 20,000 (19.8%) of men between 50 & 64 who underwent randomization had died (203)

-   1981/10,000 (19.8%) in the screening group and 1982/10,000 (19.8%) in the control group died

-    of those who died, 44/1981 (2.2%) & 78/1982 (3.9%) died from prostate cancer, respectively

  • overall the relative risk reduction in mortality was 44% for men randomized to screening compared with controls at 14 years.
  • Overall, 293 men needed to be invited for screening and 12 to be diagnosed to prevent one prostate cancer death

 

Overall, the benefits of early detection of prostate cancer increase with time.

 

Findings are based exclusively on systematic reviews (meta-analyses) of 6 randomized controlled [RCTs] PSA screening trials with 8 systematic appraisals of these RCTs but

 

  • RCTs are not the only form of evidence: absence of RCT evidence does not equal evidence of absence

 

  • These were mass population screening trials – no patient selection - as opposed to opportunistic & selective screening (which most people advocate)

 

Recently the ERSPC and PLCO data were analyzed considering implementation and practice settings between the trials, which estimated a similar effect between the trials and that screening conferred a 7-9% reduction in prostate cancer specific mortality per year and 26-31% lower risk of prostate cancer death with screening (213). This analysis has been reported to conclude that PSA screening reduced prostate cancer mortality; however the optimal screening strategy is yet to be determined or implemented to maximize benefit and reduce risk (214).  One recently proposed strategy (as discussed above) has been based on a PSA level at age 60, suggesting that men with PSA <1 ng/mL at age 60 require no further screening, while men with PSA levels ≥2 ng/mL can expect a large reduction in cancer mortality, resulting in an estimated 23 men needing to be screened and six diagnosed to avoid one prostate cancer death by 15 years (215).

 

Survival Estimation

 

There are several approaches that can be used to improve a rough clinical estimation of a patient’s life-expectancy. Validated instruments are available such as a modified form of the Total Illness Burden Index for prostate cancer by Litwin (216)and the Charlson Comorbidity Index, which seems to be most useful in men<65 years undertaking initial treatment, in particular radical prostatectomy (217-218). Although these are not used commonly in clinical practice, they do provide one option. Froehner et al (2013) recently examined available comorbidityassessments to determine which may best assist in the treatment choice for elderly men with prostate cancer. A total of 1,106 men aged 65 years or older who underwent radical prostatectomy for clinically localized prostate cancer was examined with overall survival as the study endpoint. They concluded that the American Society of Anesthesiologists (ASA) physical status classification tool, supplemented by a list of more clearly defined concomitant diseases, could be useful in clinical practice and outcome studies (219).

Another approach is to refer to Life Expectancy Tables (such as the Table 10 below modified from the Australian Bureau of Statistics website 2017). Such tables do not take into account an individual’s comorbidities.

 

Table 10: Life Expectancy Table for Australia

Age 2000-2002 2004-2006 2010-2012 2014-2016 Age 2000-2002 2004-2006 2010-2012 2014-2016
35 44.08 45.17 46.1 46.6 68 15.14 15.97 16.8 17.2
36 43.14 44.22 45.2 45.6 69 14.42 15.23 16.0 16.5
37 42.20 43.27 44.2 44.7 70 13.72 14.51 15.3 15.7
38 41.25 42.32 43.3 43.7 71 13.04 13.80 14.5 14.9
39 40.31 41.37 42.3 42.8 72 12.38 13.10 13.8 14.2
40 39.37 40.43 41.4 41.8 73 11.74 12.42 13.1 13.5
41 38.43 39.49 40.4 40.9 74 11.11 11.76 12.4 12.8
42 37.49 38.55 39.5 39.9 75 10.51 11.12 11.7 12.1
43 36.56 37.61 38.6 39.0 76 9.92 10.50 11.0 11.4
44 35.63 36.68 37.6 38.1 77 9.36 9.90 10.4 10.7
45 34.70 35.74 36.7 37.1 78 8.82 9.32 9.8 10.1
46 33.78 34.82 35.8 36.2 79 8.29 8.76 9.2 9.5
47 32.86 33.89 34.8 35.3 80 7.79 8.22 8.6 8.9
48 31.94 32.98 33.9 34.4 81 7.31 7.70 8.0 8.3
49 31.02 32.06 33.0 33.5 82 6.84 7.21 7.5 7.7
50 30.11 31.15 32.1 32.5 83 6.40 6.75 7.0 7.2
51 29.21 30.24 31.2 31.6 84 5.98 6.31 6.5 6.7
52 28.30 29.34 30.3 30.7 85 5.59 5.90 6.1 6.2
53 27.41 28.45 29.4 29.8 86 5.23 5.50 5.7 5.8
54 26.52 27.55 28.5 29.0 87 4.90 5.12 5.3 5.4
55 25.64 26.67 27.6 28.1 88 4.61 4.77 4.9 5.0
56 24.76 25.79 26.7 27.2 89 4.34 4.45 4.6 4.6
57 23.90 24.92 25.9 26.3 90 4.10 4.17 4.3 4.3
58 23.05 24.05 25.0 25.5 91 3.89 3.92 4.0 4.0
59 22.20 23.20 24.1 24.6 92 3.69 3.71 3.8 3.7
60 21.37 22.35 23.3 23.8 93 3.51 3.53 3.5 3.5
61 20.55 21.51 22.4 22.9 94 3.34 3.37 3.3 3.2
62 19.73 20.69 21.6 22.1 95 3.18 3.24 3.1 3.0
63 18.94 19.87 20.8 21.3 96 3.03 3.13 2.9 2.8
64 18.15 19.07 20.0 20.4 97 2.89 3.04 2.7 2.6
65 17.37 18.27 19.1 19.6 98 2.76 2.94 2.6 2.5
66 16.61 17.50 18.3 18.8 99 2.65 2.84 2.4 2.3
67 15.87 16.73 17.6 18.0

 

In terms of likelihood of dying from cardiovascular disease, whether or not a man has started to have erectile dysfunction may serve as a surrogate indicator. One recent large study indicated that the median time to death from a cardiovascular cause from the onset of erectile dysfunction (ED) was 10 years (220)since the reason for ED in the majority of cases is impaired arterial flow (221).

 

Factors To Consider When Deciding To Test For Prostate Cancer

 

  • In the Scandinavian randomized trial of Radical Prostatectomy & watchful waiting. At a median follow-up of 13.4 years, 63 in the surgery group and 99 in the watchful-waiting group died from prostate cancer; the relative risk was 0.56 (95% confidence interval [CI], 0.41 to 0.77; P=0.001). The number needed to treat to prevent one death due to prostate cancer was 8 (222)

 

  • The benefit of surgery with respect to death from prostate cancer was largest in men younger than 65 years of age (relative risk, 0.45) and in those with intermediate-risk prostate cancer (222)

 

  • At a median of 12.8 years of follow-up in an earlier report on this trial, men with more than 2 significant co-morbidities did not benefit from PSA testing (223)

 

  • In a follow up analysis of the PLCO study, there was a striking mortality benefit in men with minimal or no co-morbidities a 44% drop in prostate cancer-specific mortality and a number needed to treat of only 5. However, for men with at least one significant co-morbidity, there was no significant difference in prostate cancer mortality (224)

 

But what constitutes a significant comorbidity? “a condition or complaint either coexisting with the principal diagnosis or arising during the episode of care or attendance at a health care facility” (225).  How do you assess it?

 

  • Crawford et al chose an expanded definition that included both ‘standard’ Charlson comorbidity index conditions and hypertension (even if well controlled), diverticulosis, gallbladder disease and obesity (224)

 

  • But when the analysis was repeated using only validated measures of comorbidity (Charlson comorbidity index conditions only), there was no interaction (226)

 

  • A simple patient-reported index, a modified form of the Total Illness Burden Index modified for prostate cancer (216)vs Charlson Comorbidity Index (217-218)

 

  • The American Society of Anesthesiologists (ASA) physical status classificationhas been recommended to serve as a basis of assessing suitability for radical prostatectomy in men >65 years (219)

 

  • Onset of erectile dysfunctionmay serve as an indicator of limited life expectancy due to cardiovascular death (220-221)

 

  • Morbidity of (frequently repeated) TRUS & T/P biopsies TRUS biopsy infections in 4.5%: 48% had rectal swabs showing Ciprofloxacin resistant bacteria (165,167,169)

 

  • High over-diagnosis rate: active surveillance, where men diagnosed with low risk prostate cancer may be monitored with serial PSA and biopsies to delay or avoid treatment, may decrease the concern of over detection and over treatment

 

  • Psychosocial aspects pervade all aspects of detection & treatment.

 

Recent studies have reported psychological distress levels severe enough to meet defined criteria close to the time of diagnosis from 10% to 23% (227).  Bill-Axelson and colleagues in an eight year longitudinal study reported that although extreme distress was not common in men with localized prostate cancer, 30–40% of men reported ongoing health-related distress and worry about their health, feeling low, and sleep disturbance (228). Risk of suicide may be increased in the first six to twelve months after the diagnosis of prostate cancer (229-230).  Screening for distress and referral to appropriate support services is widely accepted and recommended in men diagnosed with prostate cancer (231-232). In addition to distress, contemporary evidence suggests socio-demographic and psychosocial variables to be highly influential on intervention effects (232). Decisional conflicts impact upon continuation of Active Surveillance (233-234). When making decisions about treatment for prostate cancer men tend to rely on lay beliefs about cancer with the opinion of the clinician highly influential (233). A systematic review of psychosocial interventions for men with prostate cancer and their partners found that group cognitive-behavioral and psychoeducational interventions were helpful in promoting better psychological adjustment and quality of life (QOL) for men with prostate cancer (235). Multi-modal psychosexual and psychosocial interventions for men diagnosed with prostate cancer are recommended(232).

 

  • Reassurance: PSA level <1ng/ml at the age of 65 years (50)or <3 ng/ml at the age of 75 years have a very low chance of contracting fatal cancer (160)

 

 

EARLY DIAGNOSIS AND TREATMENT WITH CURATIVE INTENET AND LESSENING THE LIKELIHOOD OF METASTASES OCCURRING

 

The recently completed PSA Evaluation Report by the National Health and Medical Research Council (NHMRC) of Australia concluded that, although there was some inconsistency in the definition of prostate cancer metastases across the RCTs, overall, the evidence indicates that PSA testing reduces the risk of having metastases present at the time of diagnosis of prostate cancer. The NHMRC review focused on the RCTs above in its considerations but did not conclude that intervention with curative intent reduces the likelihood of subsequent metastases [163].  However, evaluation of evidence from multiple non-RCTs has reported that PSA testing and intervention with curative intent does reduce the likelihood of subsequent metastases.

 

There are very few RCTs for prostate cancer treated with curative intent. Bill Axelson et al (2014) (222)recruited patients from 14 centers in Sweden, Finland and Iceland: The trial is noteworthy since the study included patients detected with prostate cancer at a later stage than is currently diagnosed: only 12% had impalpable disease on DRE - detected by what are now outmoded methods. The results are summarized below:

 

Swedish Trial Of Radical Prostatectomy Versus Watchful Waiting (222,236)

 

  • From October 1989 through February 1999, 695 men with ‘early’ prostate cancer were randomly assigned to watchful waiting, where men are “watched” and treated only when symptomatic or with significant concern for complications, or radical prostatectomy
  • Eligibility required patients to be
  • <75 yrs. of age and a life expectancy >10 years: mean age was 65 yrs.
  • Clinically localized disease (T1 or T2, using IUCC 1978 criteria)
  • Diagnosis by core biopsy or fine needle aspiration cytology
  • Well or moderately differentiated adenocarcinoma (WHO classification)
  • PSA <50 ng/ml: mean PSA was 13 ng/ml
  • a negative bone scan
  • During a median of 13.4 years, 200 of the 347 men in the radical prostatectomy group and 247 of the 348 in the watchful-waiting group died (222). In the case of 63 men assigned to surgery and 99 men assigned to watchful waiting, death was due to prostate cancer (P = 0.001)
  • The survival benefit was largest in men younger than 65 years of age and those with intermediate-risk prostate cancer
  • The number needed to treat to avert one death at 18 years of follow-up was 8 (P=0.001) and 4 for men younger than 65 years of age (222)
  • Among men who underwent radical prostatectomy, those with extracapsular tumor growth had a risk of death from prostate cancer that was 7 times that of men without extracapsular tumor growth (236)
  • Distant metastases were diagnosed in 89 men in the radical prostatectomy group and 138 in the watchful waiting cohort resulting in a relative risk of metastases in the RP group of 0.57 (P <0.001) (222)

 

However, by contrast, in the Prostate Cancer Intervention versus Observation Trial (PIVOT) of radicalprostatectomy versus observation for localized prostate cancer found differently (237). Between November 1994 and January 2002, 731 men with localized prostate cancer (mean age, 67 years; median PSA value, 7.8 ng per milliliter) were randomly assigned to radical prostatectomy or observation and followed to January 2010. The primary outcome was all-cause mortality; the secondary outcome was prostate-cancer mortality

 

During the median follow-up of 10.0 years, 171 of 364 men (47.0%) assigned to radical prostatectomy died, compared with 183 of 367 (49.9%) assigned to observation (P=0.22). Among men assigned to radical prostatectomy, 21 (5.8%) died from prostate cancer or treatment, compared with 31 men (8.4%) assigned to observation (P=0.09). The effect of treatment on all-cause and prostate-cancer mortality did not differ according to age, race, coexisting conditions, self-reported performance status, or histological features of the tumor. Radical prostatectomy was associated with reduced all-cause mortality among men with a PSA value greater than 10 ng per milliliter (P=0.04 for interaction) and possibly among those with intermediate-risk or high-risk tumors (P=0.07 for interaction). Adverse events within 30 days after surgery occurred in 21.4% of men, including one death. In 2017, Wilt et al updated the results reporting that after nearly 20 years of follow-up among men with localized prostate cancer: surgery, was not associated with significantly lower all-cause or prostate-cancer mortality than observation.

 

However, there were serious deficiencies with the PIVOT study (238). Although the three Endpoints Committee members were blinded to randomized treatment assignments, reviewed medical records and death certificates when available to assign a cause of death using a primary and a secondary adjudication question, initial disagreements were resolved through discussion. Complete agreement on cause of death by all three committee members before any discussion was achieved in 200/354 (56%) cases on the primary and 209/354 (59%) cases on the secondary. Complete agreement on the primary cause rose to 306/354 (86%) when ‘definite’ and ‘probably’ categories were collapsed, as planned a priori. There was no separate ‘gold standard’ by which to judge the accuracy of the final endpoints committee adjudications, and useful death certificates could not be obtained on about a third of PIVOT participants who died.

 

U.S. PIVOT – Radical Prostatectomy Versus Observation (237)

 

  • Recruitment difficulties and patient compliance issues affected numbers so that only 731 of the proposed 2000 men could be recruited to the trial and hence this study is considered to be underpowered to detect a difference in overall survival (239)
  • Serious lack of agreement on cause of death by Endpoint Committee Members: Useful death certificates could not be obtained for approximately one third of participants
  • Differences between histological reporting at participating sites and by a central pathologist affected risk stratification and, consequently, secondary endpoint results
  • A less predictive pre-2005 ISUP Consensus Gleason classification was used with ~25% of patients with Gleason scores of 7 or higher reported at the peripheral sites compared with 48% with Gleason scores 7 or higher by a central pathologist

 

Consequently, the answer based on RCT evidence remains uncertain.

 

 

Early Diagnosis And Treatment With Curative Intent: Avoiding The Late Clinical Problems Resulting From A Large Pelvic Tumor

 

There is a paucity of high level evidence that early diagnosis of prostate cancer will prevent or minimize the problems resulting from a large pelvic tumor. Anecdotally, managing patients with disabling symptoms from advanced local prostate cancer constituted a considerable part of a urologist’s workload. Frequent visits to hospital for interventions together with burden of clinical symptoms such as unremitting day and night frequency, incontinence and bleeding, impact significantly on the dignity and quality of life of these men (240).

Figure 17: CT of pelvis showing prostatic tumor extending into the (thick-walled) bladder and spread to involve pelvic lymph nodes: the patient had multiple lower urinary tract symptoms

WHETHER TO TEST FOR PROSTATE CANCER

 

Prostate cancer is the most common male visceral malignancy in the developed world and the second most common cause of cancer deaths, uncertainties remain about management practices at several points in the illness continuum. For example, owing to controversies regarding the outcomes of screening trials for prostate cancer reducing the death rate from this disease, population-based screening for prostate cancer in asymptomatic men is not currently recommended in most countries (241).  Rather, it is suggested that men should be able to access PSA testing as long as they are fully informed of the pros and cons of testing.

The Prostate Cancer Foundation of Australia (PCFA), in partnership with National Health and Medical Research Council (NHMRC) and Cancer Council Australia, published a “PSA Testing for Prostate Cancer in Asymptomatic Men” guideline in 2016, which was commissioned by the Department of Health and comprised a multi-disciplinary expert advisory panel. The guidelines have been endorsed by the Urological Society of Australia and New Zealand (USANZ) and the Royal Australian College of General Practitioners (RACGP). The recommendations are as follows:

 

  • A population screening program for prostate cancer (a program that offers testing to all men of a certain age group) is not recommended
  • Men should be offered evidence-based decision support, including the opportunity to discuss the benefits and harms of PSA testing before making the decision to be tested
  • Men at average risk of prostate cancer who decide to be tested should be offered PSA testing every 2 years from age 50 to 69
  • The harms of PSA testing may outweigh the benefits for men aged 70 and older or those with a life expectancy less than 7 years.
  • Men with a family history of prostate cancer who decide to be tested should be offered PSA testing every 2 years from age 40/ 45 to 69 with the starting age depending on the strength of their family history
  • Digital rectal examination is not recommended in addition to PSA testing in the primary care setting

 

The full guideline can be accessed at www.pcfa.org.au/psa-testing-guidelinesor https://wiki.cancer.org.au/australia/Guidelines:PSA_Testing

 

The approach that is considered to be optimal for achieving high quality patient decisions is shared decision making (242).

 

Shared decision makingis defined as a process carried out between a patient and his health care professional where both parties share information and the patient understands the risks and benefits of each treatment option, participates in the decision to the extent that he desires and makes a decision consistent with his preferences and values, or defers the decision to another time(243).

 

Shared decision making may not be easy to achieve for all patients (243).  For example, although many patients with cancer indicate a preference for sharing decision making with their clinicians, some, in the case of prostate cancer between 8% to 58% of men, prefer a passive decision-making role where clinicians make treatment decisions on their behalf   (244-245).  However, clinicians still need to understand patients’ preferences to ensure that they are making quality decisions on behalf of their patients. As well, there is often a gap between the clinical ideal of shared decision making and actual clinical practice where decision complexity and time constraints may make this approach difficult for both parties to achieve (246-247).  There are, however, defined strategies and decision aids that can facilitate this process (248).

 

Supporting Patient Choice About Testing For Prostate Cancer

 

Many groups advocate an informed decision-making process as an evidence-based approach and necessary precursor to screening for early prostate cancer (241,249).  Others have suggested that informed decision-making on this health topic is also necessary as a medico-legal risk management strategy (250-251).  While some researchers have suggested a set of information that needs to be communicated to men about this health decision (252-253), there are few explicit guidelines on this subject (254).  Problematically, patients and clinicians do not agree on core content, including a basic explanation of a PSA test and the psychological effects of a positive PSA test result (255).  It has been advised that, for any screening test, patients need to understand the purpose of the test, the likelihood of false-negatives and false-positives, the uncertainties and risks associated with testing, significant medical, social or financial implications of testing and any possible sequelae and follow up care plans (256)www.ipdas.ohri.ca.

 

Such information needs to be communicated to patients in a logical and balanced sequence in order to promote better understanding and increased decisional control by men.  One approach that has been proposed in primary care in Australia is the use of six decision steps (see Figure 18).  Each decision step logically follows to prompt the clinician to overview important health information, with tailoring suggested in Step 1 to ensure the discussion is consistent with the patient’s concerns.  For example, for a man with a significant family history of prostate cancer, this factor is likely to be central to the patient discussion (257).  Men who experience uncomplicated LUTS often worry about prostate cancer, so addressing this concern first may be priority (258-259).  In this regard, resources for patients that explain about male reproductive health problems such as urinary symptoms and sexual dysfunction are available at www.andrologyaustralia.org. As well, National Health and Medical Research Council guidelines are available about the management of LUTS http://www.health.gov.au/nhmrc/publications/synopses/cp42syn.htm

 

Other overseas websites include:

Figure 18: Six Decision Steps

From this point, checking to ensure the patient has a basic understanding of both the prostate and possible tests is needed and, given many men may be unaware of the location and function of the prostate gland, an anatomical diagram may be a useful teaching tool here.  Next, a consideration of individual risk with regard to both the incidence and mortality of prostate cancer is needed.  Communicating health risks effectively is a challenge in the provision of effective decision support.  In general people find probabilities hard to understand, often estimate their level of risk incorrectly, and tend not to weigh up pros and cons in a systematic way when deciding about treatments (234,260-261).  As well, population-based statistics provide data about populations, not individuals, so risk communication needs to acknowledge this as a limitation and, where possible, refer to age-based risk estimates and relevant individual factors such as family history) (262).

 

There are a number of communication strategies that have been suggested to help patients understand risk.  These include

  • using numbers as well as words to explain risk
  • where possible providing the absolute risk or benefit
  • using frequencies rather than single event probabilities
  • using consistent denominators
  • putting the risk into context by comparing it to other life events
  • offering both the possible negative and positive outcomes to balance the message frame (263-265).

 

However, a quality health decision goes beyond the simple transfer of information and includes consideration and incorporation of each patient’s values and personal preferences (266).  Thus, Step 5 in Box 1 prompts the clinician to discuss each man’s individual preferences. A number of strategies can be used to do this, most commonly the use of a pros and cons exercise in which patients are encouraged to explicitly consider the factors that matter most to them personally in this decision, and the direction and leaning of their preferences either for or against each possible option.  One approach to support this process for this health topic is the inclusion of a values table within a decision card (see Table 11).  A decision aid that incorporates both the six decision steps and this values clarification exercise can be found on the Andrology Australia website at: http://www.prostate.org.au/articleLive/attachments/1/GP%20Show%20Card%20041007.pdf

 

Table 11:  What is most important to you?

FOR: Is this like you? AGAINST: Is this like you?

 

I’m concerned that I might get prostate cancer I think my chance of getting prostate cancer is low
I want the best chance of finding it early, if I do get it I am not convinced about the effectiveness of testing
I’m not interested in waiting for all the proof to be in I am more concerned about avoiding treatment side effects, if there’s no guarantee I’d be reducing my risk of dying from prostate cancer
I want to do everything possible to reduce my risk of dying from prostate cancer

 

Decision aids are also effective in supporting patients to make informed choices.  With regards to PSA testing, patient-focused decision aids and decision counselling or support interventions have been found to be effective in increasing men’s knowledge about PSA testing and decreasing decision-related distress (254,267-270), with a variable effect on actual testing behavior.

 

A range of aids is freely available from the web (www.prostatehealth.org.au; www.cdc.gov/cancer/prostate;www.cancerbacup.org.uk). 

 

Cancer helplines also often provide such information, for example:

  • The Cancer Council Australia Cancer Helpline on 13 11 20;
  • the UK helpline on 0808 800 1234;
  • the USA Cancer Helpline on 1800 227 2345.

 

REFERENCES

 

  1. Surveillance E, and End Results Program (SEER). Prostate Cancer - Cancer Stat Facts.http://seer.cancer.gov/statfacts/html/prost.html.
  2. Siegel R, Naishadham D, Jemal A. Cancer statistics, 2012. CA Cancer J Clin2012; 62:10-29
  3. Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F. Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. International Journal of Cancer2015; 136:E359-E386
  4. Baade PD, Youlden DR, Cramb SM, Dunn J, Gardiner RA. Epidemiology of prostate cancer in the Asia-Pacific region. Prostate international2013; 1:47-58
  5. Center MM, Jemal A, Lortet-Tieulent J, Ward E, Ferlay J, Brawley O, Bray F. International Variation in Prostate Cancer Incidence and Mortality Rates. European Urology2012; 61:1079-1092
  6. AIHW National Mortality Database. 2013; http://www.aihw.gov.au/cancer.
  7. Malvezzi M, Bertuccio P, Levi F, La Vecchia C, Negri E. European cancer mortality predictions for the year 2013. Annals of Oncology2013;
  8. Welfare AIoH. Prostate Cancer in Australia. Canberra: AIHW;2013.
  9. Baade PD, Coory MD, Aitken JF. International trends in prostate-cancer mortality: the decrease is continuing and spreading. Cancer causes & control : CCC2004; 15:237-241
  10. Feletto E, Bang A, Cole-Clark D, Chalasani V, Rasiah K, Smith DP. An examination of prostate cancer trends in Australia, England, Canada and USA: Is the Australian death rate too high? World Journal of Urology2015; 33:1677-1687
  11. Bartsch G, Horninger W, Klocker H, Reissigl A, Oberaigner W, Schonitzer D, Severi G, Robertson C, Boyle P. Prostate cancer mortality after introduction of prostate-specific antigen mass screening in the Federal State of Tyrol, Austria. Urology2001; 58:417-424
  12. Etzioni R, Legler JM, Feuer EJ, Merrill RM, Cronin KA, Hankey BF. Cancer surveillance series: interpreting trends in prostate cancer--part III: Quantifying the link between population prostate-specific antigen testing and recent declines in prostate cancer mortality. Journal of the National Cancer Institute1999; 91:1033-1039
  13. Hankey BF, Feuer EJ, Clegg LX, Hayes RB, Legler JM, Prorok PC, Ries LA, Merrill RM, Kaplan RS. Cancer surveillance series: interpreting trends in prostate cancer--part I: Evidence of the effects of screening in recent prostate cancer incidence, mortality, and survival rates. Journal of the National Cancer Institute1999; 91:1017-1024
  14. SchröderFH, Kranse R. Verification Bias and the Prostate-Specific Antigen Test — Is There a Case for a Lower Threshold for Biopsy? New England Journal of Medicine2003; 349:393-395
  15. Partin AW, Stutzman RE. Elevated prostate-specific antigen, abnormal prostate evaluation on digital rectal examination, and transrectal ultrasound and prostate biopsy. The Urologic clinics of North America1998; 25:581-589, viii
  16. Jhaveri FM, Klein EA, Kupelian PA, Zippe C, Levin HS. Declining rates of extracapsular extension after radical prostatectomy: evidence for continued stage migration. Journal of clinical oncology : official journal of the American Society of Clinical Oncology1999; 17:3167-3172
  17. Brawer MK. Prostate-specific antigen: Critical issues. Urology1994; 44:9-17
  18. Carter HB, Pearson JD. Prostate-specific antigen testing for early diagnosis of prostate cancer: formulation of guidelines. Urology54:780-786
  19. Gann PH, Hennekens CH, Stampfer MJ. A prospective evaluation of plasma prostate-specific antigen for detection of prostatic cancer. Jama1995; 273:289-294
  20. Smith DS, Catalona WJ. The nature of prostate cancer detected through prostate specific antigen based screening. The Journal of urology1994; 152:1732-1736
  21. Hoedemaeker RF, Rietbergen JB, Kranse R, Schroder FH, van der Kwast TH. Histopathological prostate cancer characteristics at radical prostatectomy after population based screening. The Journal of urology2000; 164:411-415
  22. Luboldt HJ, Bex A, Swoboda A, Husing J, Rubben H. Early detection of prostate cancer in Germany: a study using digital rectal examination and 4.0 ng/ml prostate-specific antigen as cutoff. Eur Urol2001; 39:131-137
  23. Freedland SJ, Presti JC, Jr., Amling CL, Kane CJ, Aronson WJ, Dorey F, Terris MK. Time trends in biochemical recurrence after radical prostatectomy: results of the SEARCH database. Urology2003; 61:736-741
  24. Labrie F, Candas B, Dupont A, Cusan L, Gomez JL, Suburu RE, Diamond P, Levesque J, Belanger A. Screening decreases prostate cancer death: first analysis of the 1988 Quebec prospective randomized controlled trial. The Prostate1999; 38:83-91
  25. Kirby R, Christmas T, Brawer MK. Prostate Cancer.London: Times Mirror International Publishers.
  26. Sakr WA, Grignon DJ, Crissman JD, Heilbrun LK, Cassin BJ, Pontes JJ, Haas GP. High grade prostatic intraepithelial neoplasia (HGPIN) and prostatic adenocarcinoma between the ages of 20-69: an autopsy study of 249 cases. In vivo (Athens, Greece)1994; 8:439-443
  27. Deaths, Life expectancy. Reports & Statisticshttps://www.aihw.gov.au/reports/life-expectancy-death/deaths-in-australia/contents/life-expectancy, 2017.
  28. Heart, stroke & vascular diseases. Reports & Statisticshttps://www.aihw.gov.au/reports-statistics/health-conditions-disability-deaths/heart-stroke-vascular-diseases/overview, 2017.
  29. Nelson Q, Agarwal N, Stephenson R, Cannon-Albright LA. A population-based analysis of clustering identifies a strong genetic contribution to lethal prostate cancer. Frontiers in Genetics2013; 4:152
  30. Steinberg GD, Carter BS, Beaty TH, Childs B, Walsh PC. Family history and the risk of prostate cancer. The Prostate1990; 17:337-347
  31. Carter BS, Bova GS, Beaty TH, Steinberg GD, Childs B, Isaacs WB, Walsh PC. Hereditary prostate cancer: epidemiologic and clinical features. The Journal of urology1993; 150:797-802
  32. Brandt A, Bermejo JL, Sundquist J, Hemminki K. Age-specific risk of incident prostate cancer and risk of death from prostate cancer defined by the number of affected family members. Eur Urol2010; 58:275-280
  33. Grönberg H, Damber L, Damber JE. Familial prostate cancer in sweden: A nationwide register cohort study. Cancer1996; 77:138-143
  34. Teerlink CC, Thibodeau SN, McDonnell SK, Schaid DJ, Rinckleb A, Maier C, Vogel W, Cancel-Tassin G, Egrot C, Cussenot O, Foulkes WD, Giles GG, Hopper JL, Severi G, Eeles R, Easton D, Kote-Jarai Z, Guy M, Cooney KA, Ray AM, Zuhlke KA, Lange EM, FitzGerald LM, Stanford JL, Ostrander EA, Wiley KE, Isaacs SD, Walsh PC, Isaacs WB, Wahlfors T, Tammela T, Schleutker J, Wiklund F, Grönberg H, Emanuelsson M, Carpten J, Bailey-Wilson J, Whittemore AS, Oakley-Girvan I, Hsieh C-L, Catalona WJ, Zheng SL, Jin G, Lu L, Xu J, Camp NJ, Cannon-Albright LA. Association analysis of 9,560 prostate cancer cases from the International Consortium of Prostate Cancer Genetics confirms the role of reported prostate cancer associated SNPs for familial disease. Human Genetics2014; 133:347-356
  35. Eeles R, Goh C, Castro E, Bancroft E, Guy M, Olama AAA, Easton D, Kote-Jarai Z. The genetic epidemiology of prostate cancer and its clinical implications. Nature Reviews Urology2013; 11:18
  36. Bratt O. What should a urologist know about hereditary predisposition to prostate cancer? BJU international2007; 99:743-747; discussion 747-748
  37. Lynch HT, Kosoko-Lasaki O, Leslie SW, Rendell M, Shaw T, Snyder C, D'Amico AV, Buxbaum S, Isaacs WB, Loeb S, Moul JW, Powell I. Screening for familial and hereditary prostate cancer. International Journal of Cancer2016; 138:2579-2591
  38. Agalliu I, Karlins E, Kwon EM, Iwasaki LM, Diamond A, Ostrander EA, Stanford JL. Rare germline mutations in the BRCA2 gene are associated with early-onset prostate cancer. British Journal of Cancer2007; 97:826-831
  39. Ford D, Easton DF, Bishop DT, Narod SA, Goldgar DE. Risks of cancer in BRCA1-mutation carriers. Breast Cancer Linkage Consortium. Lancet (London, England)1994; 343:692-695
  40. Thompson D, Easton DF. Cancer Incidence in BRCA1 mutation carriers. Journal of the National Cancer Institute2002; 94:1358-1365
  41. Dobson R. Prostate cancer patients with BRCA2 mutation face poor survival. BMJ (Clinical research ed)2008; 337:a705
  42. Tryggvadottir L, Vidarsdottir L, Thorgeirsson T, Jonasson JG, Olafsdottir EJ, Olafsdottir GH, Rafnar T, Thorlacius S, Jonsson E, Eyfjord JE, Tulinius H. Prostate cancer progression and survival in BRCA2 mutation carriers. Journal of the National Cancer Institute2007; 99:929-935
  43. Narod SA, Neuhausen S, Vichodez G, Armel S, Lynch HT, Ghadirian P, Cummings S, Olopade O, Stoppa-Lyonnet D, Couch F, Wagner T, Warner E, Foulkes WD, Saal H, Weitzel J, Tulman A, Poll A, Nam R, Sun P, Danquah J, Domchek S, Tung N, Ainsworth P, Horsman D, Kim-Sing C, Maugard C, Eisen A, Daly M, McKinnon W, Wood M, Isaacs C, Gilchrist D, Karlan B, Nedelcu R, Meschino W, Garber J, Pasini B, Manoukian S, Bellati C. Rapid progression of prostate cancer in men with a BRCA2 mutation. Br J Cancer2008; 99:371-374
  44. Castro E, Goh C, Olmos D, Saunders E, Leongamornlert D, Tymrakiewicz M, Mahmud N, Dadaev T, Govindasami K, Guy M, Sawyer E, Wilkinson R, Ardern-Jones A, Ellis S, Frost D, Peock S, Evans DG, Tischkowitz M, Cole T, Davidson R, Eccles D, Brewer C, Douglas F, Porteous ME, Donaldson A, Dorkins H, Izatt L, Cook J, Hodgson S, Kennedy MJ, Side LE, Eason J, Murray A, Antoniou AC, Easton DF, Kote-Jarai Z, Eeles R. Germline BRCA Mutations Are Associated With Higher Risk of Nodal Involvement, Distant Metastasis, and Poor Survival Outcomes in Prostate Cancer. Journal of Clinical Oncology2013; 31:1748-1757
  45. Bancroft EK, Page EC, Castro E, Lilja H, Vickers A, Sjoberg D, Assel M, Foster CS, Mitchell G, Drew K, Mæhle L, Axcrona K, Evans DG, Bulman B, Eccles D, McBride D, van Asperen C, Vasen H, Kiemeney LA, Ringelberg J, Cybulski C, Wokolorczyk D, Selkirk C, Hulick PJ, Bojesen A, Skytte A-B, Lam J, Taylor L, Oldenburg R, Cremers R, Verhaegh G, van Zelst-Stams WA, Oosterwijk JC, Blanco I, Salinas M, Cook J, Rosario DJ, Buys S, Conner T, Ausems MG, Ong K-r, Hoffman J, Domchek S, Powers J, Teixeira MR, Maia S, Foulkes WD, Taherian N, Ruijs M, den Enden ATH-v, Izatt L, Davidson R, Adank MA, Walker L, Schmutzler R, Tucker K, Kirk J, Hodgson S, Harris M, Douglas F, Lindeman GJ, Zgajnar J, Tischkowitz M, Clowes VE, Susman R, Ramón y Cajal T, Patcher N, Gadea N, Spigelman A, van Os T, Liljegren A, Side L, Brewer C, Brady AF, Donaldson A, Stefansdottir V, Friedman E, Chen-Shtoyerman R, Amor DJ, Copakova L, Barwell J, Giri VN, Murthy V, Nicolai N, Teo S-H, Greenhalgh L, Strom S, Henderson A, McGrath J, Gallagher D, Aaronson N, Ardern-Jones A, Bangma C, Dearnaley D, Costello P, Eyfjord J, Rothwell J, Falconer A, Gronberg H, Hamdy FC, Johannsson O, Khoo V, Kote-Jarai Z, Lubinski J, Axcrona U, Melia J, McKinley J, Mitra AV, Moynihan C, Rennert G, Suri M, Wilson P, Killick E, Moss S, Eeles RA. Targeted Prostate Cancer Screening in BRCA1 and BRCA2 Mutation Carriers: Results from the Initial Screening Round of the IMPACT Study. European Urology2014; 66:489-499
  46. Constantinou J, Feneley MR. PSA testing: an evolving relationship with prostate cancer screening. Prostate cancer and prostatic diseases2006; 9:6-13
  47. Thompson IM, Ankerst DP, Chi C, Lucia MS, Goodman PJ, Crowley JJ, Parnes HL, Coltman CA, Jr. Operating characteristics of prostate-specific antigen in men with an initial PSA level of 3.0 ng/ml or lower. Jama2005; 294:66-70
  48. Schröder FH, Hugosson J, Roobol MJ, Tammela TL, Ciatto S, Nelen V, Kwiatkowski M, Lujan M, Lilja H, Zappa M, Denis LJ, Recker F, Berenguer A, Määttänen L, Bangma CH, Aus G, Villers A, Rebillard X, van der Kwast T, Blijenberg BG, Moss SM, de Koning HJ, Auvinen A. Screening and prostate-cancer mortality in a randomized European study. N Engl J Med2009; 360:1320-1328
  49. Lodding P, Aus G, Bergdahl S, Frosing R, Lilja H, Pihl CG, Hugosson J. Characteristics of screening detected prostate cancer in men 50 to 66 years old with 3 to 4 ng./ml. Prostate specific antigen. The Journal of urology1998; 159:899-903
  50. Vickers AJ, Cronin AM, Bjork T, Manjer J, Nilsson PM, Dahlin A, Bjartell A, Scardino PT, Ulmert D, Lilja H. Prostate specific antigen concentration at age 60 and death or metastasis from prostate cancer: case-control study. BMJ (Clinical research ed)2010; 341:c4521
  51. Marks LS, Andriole GL, Fitzpatrick JM, Schulman CC, Roehrborn CG. The interpretation of serum prostate specific antigen in men receiving 5alpha-reductase inhibitors: a review and clinical recommendations. The Journal of urology2006; 176:868-874
  52. Giganti F, Moore CM, Robertson NL, McCartan N, Jameson C, Bott SRJ, Winkler M, Gambarota G, Whitcher B, Castro R, Emberton M, Allen C, Kirkham A. MRI findings in men on active surveillance for prostate cancer: does dutasteride make MRI visible lesions less conspicuous? Results from a placebo-controlled, randomised clinical trial. European Radiology2017; 27:4767-4774
  53. Thompson IM, Jr., Goodman PJ, Tangen CM, Parnes HL, Minasian LM, Godley PA, Lucia MS, Ford LG. Long-term survival of participants in the prostate cancer prevention trial. N Engl J Med2013; 369:603-610
  54. Ornstein DK, Smith DS, Humphrey PA, Catalona WJ. The effect of prostate volume, age, total prostate specific antigen level and acute inflammation on the percentage of free serum prostate specific antigen levels in men without clinically detectable prostate cancer. The Journal of urology1998; 159:1234-1237
  55. Yuan JJ, Coplen DE, Petros JA, Figenshau RS, Ratliff TL, Smith DS, Catalona WJ. Effects of rectal examination, prostatic massage, ultrasonography and needle biopsy on serum prostate specific antigen levels. The Journal of urology1992; 147:810-814
  56. Kang D-Y, Li H-J. The Effect of Testosterone Replacement Therapy on Prostate-Specific Antigen (PSA) Levels in Men Being Treated for Hypogonadism: A Systematic Review and Meta-Analysis. Medicine2015; 94:e410
  57. Boyle P, Koechlin A, Bota M, d'Onofrio A, Zaridze DG, Perrin P, Fitzpatrick J, Burnett AL, Boniol M. Endogenous and exogenous testosterone and the risk of prostate cancer and increased prostate-specific antigen (PSA) level: a meta-analysis. BJU international2016; 118:731-741
  58. Dupree JM, Langille GM, Khera M, Lipshultz LI. The safety of testosterone supplementation therapy in prostate cancer. Nature Reviews Urology2014; 11:526
  59. Pearl JA, Berhanu D, François N, Masson P, Zargaroff S, Cashy J, McVary KT. Testosterone Supplementation does not Worsen Lower Urinary Tract Symptoms. The Journal of urology2013; 190:1828-1833
  60. Rider JR, Wilson KM, Sinnott JA, Kelly RS, Mucci LA, Giovannucci EL. Ejaculation Frequency and Risk of Prostate Cancer: Updated Results with an Additional Decade of Follow-up. European Urology2016; 70:974-982
  61. Morgan TO, Jacobsen SJ, McCarthy WF, Jacobson DJ, McLeod DG, Moul JW. Age-Specific Reference Ranges for Serum Prostate-Specific Antigen in Black Men. New England Journal of Medicine1996; 335:304-310
  62. DeAntoni EP, Crawford ED, Oesterling JE, Ross CA, Berger ER, McLeod DG, Staggers F, Stone NN. Age- and race-specific reference ranges for prostate-specific antigen from a large community-based study. Urology1996; 48:234-239
  63. Mottet N, Bellmunt J, Bolla M, Briers E, Cumberbatch MG, De Santis M, Fossati N, Gross T, Henry AM, Joniau S, Lam TB, Mason MD, Matveev VB, Moldovan PC, van den Bergh RC, Van den Broeck T, van der Poel HG, van der Kwast TH, Rouviere O, Schoots IG, Wiegel T, Cornford P. EAU-ESTRO-SIOG Guidelines on Prostate Cancer. Part 1: Screening, Diagnosis, and Local Treatment with Curative Intent. Eur Urol2017; 71:618-629
  64. Oesterling JE, Jacobsen SJ, Klee GG, Pettersson K, Piironen T, Abrahamsson PA, Stenman UH, Dowell B, Lovgren T, Lilja H. Free, complexed and total serum prostate specific antigen: the establishment of appropriate reference ranges for their concentrations and ratios. The Journal of urology1995; 154:1090-1095
  65. Fang J, Metter EJ, Landis P, Chan DW, Morrell CH, Carter HB. Low levels of prostate-specific antigen predict long-term risk of prostate cancer: results from the Baltimore Longitudinal Study of Aging. Urology2001; 58:411-416
  66. Carter HB, Pearson JD, Metter EJ, Brant LJ, Chan DW, Andres R, Fozard JL, Walsh PC. Longitudinal evaluation of prostate-specific antigen levels in men with and without prostate disease. Jama1992; 267:2215-2220
  67. Ku JH. Race-specific reference ranges of serum prostate-specific antigen levels in countries with a low incidence of prostate cancer. BJU international2006; 97:69-72
  68. Imai K, Ichinose Y, Kubota Y, Yamanaka H, Sato J, Saitoh M, Watanabe H, Ohe H. Clinical significance of prostate specific antigen for early stage prostate cancer detection. Japanese journal of clinical oncology1994; 24:160-165
  69. Imai K, Ichinose Y, Kubota Y, Yamanaka H, Sato J. Diagnostic significance of prostate specific antigen and the development of a mass screening system for prostate cancer. The Journal of urology1995; 154:1085-1089
  70. Ito K, Yamamoto T, Kubota Y, Suzuki K, Fukabori Y, Kurokawa K, Yamanaka H. Usefulness of age-specific reference range of prostate-specific antigen for Japanese men older than 60 years in mass screening for prostate cancer. Urology2000; 56:278-282
  71. He D, Wang M, Chen X, Gao Z, He H, Zhau HE, Wang W, Chung LW, Nan X. Ethnic differences in distribution of serum prostate-specific antigen: a study in a healthy Chinese male population. Urology2004; 63:722-726
  72. Lin WY, Gu CJ, Kao CH, Changlai SP, Wang SJ. Serum prostate-specific antigen in healthy Chinese men: establishment of age-specific reference ranges. Neoplasma1996; 43:103-105
  73. Kao CH. Age-related free PSA, total PSA and free PSA/total PSA ratios: establishment of reference ranges in Chinese males. Anticancer research1997; 17:1361-1365
  74. Wu TT, Huang JK. The clinical usefulness of prostate-specific antigen (PSA) level and age-specific PSA reference ranges for detecting prostate cancer in Chinese. Urologia internationalis2004; 72:208-211
  75. Tay KP, Chin CM, Lim PH, Chng HC. Prostate screening--the Singapore experience. International journal of urology : official journal of the Japanese Urological Association1996; 3:102-107
  76. Saw S, Aw TC. Age-related reference intervals for free and total prostate-specific antigen in a Singaporean population. Pathology2000; 32:245-249
  77. Lee SE, Kwak C, Park MS, Lee CH, Kang W, Oh SJ. Ethnic differences in the age-related distribution of serum prostate-specific antigen values: a study in a healthy Korean male population. Urology2000; 56:1007-1010
  78. Ku JH, Ahn JO, Lee CH, Lee NK, Park YH, Byun SS, Kwak C, Lee SE. Distribution of serum prostate-specific antigen in healthy Korean men: influence of ethnicity. Urology2002; 60:475-479
  79. Roobol MJ, Schröder FH, Crawford ED, Freedland SJ, Sartor AO, Fleshner N, Andriole GL. A Framework for the Identification of Men at Increased Risk for Prostate Cancer. The Journal of urology2009; 182:2112-2122
  80. Thompson IM, Tangen CM, Ankerst DP, Chi C, Lucia MS, Goodman P, Parnes H, Coltman CA, Jr. The performance of prostate specific antigen for predicting prostate cancer is maintained after a prior negative prostate biopsy. The Journal of urology2008; 180:544-547
  81. Pelzer AE, Tewari A, Bektic J, Berger AP, Frauscher F, Bartsch G, Horninger W. Detection rates and biologic significance of prostate cancer with PSA less than 4.0 ng/mL: observation and clinical implications from Tyrol screening project. Urology2005; 66:1029-1033
  82. Vickers AJ, Ulmert D, Sjoberg DD, Bennette CJ, Bjork T, Gerdtsson A, Manjer J, Nilsson PM, Dahlin A, Bjartell A, Scardino PT, Lilja H. Strategy for detection of prostate cancer based on relation between prostate specific antigen at age 40-55 and long term risk of metastasis: case-control study. BMJ (Clinical research ed)2013; 346:f2023
  83. Aus G, Damber JE, Khatami A, Lilja H, Stranne J, Hugosson J. Individualized screening interval for prostate cancer based on prostate-specific antigen level: results of a prospective, randomized, population-based study. Archives of internal medicine2005; 165:1857-1861
  84. Carter HB, Pearson JD. PSA velocity for the diagnosis of early prostate cancer. A new concept. The Urologic clinics of North America1993; 20:665-670
  85. Makarov DV, Loeb S, Magheli A, Zhao K, Humphreys E, Gonzalgo ML, Partin AW, Han M. Significance of preoperative PSA velocity in men with low serum PSA and normal DRE. World J Urol2011; 29:11-14
  86. D'Amico AV, Chen MH, Roehl KA, Catalona WJ. Preoperative PSA velocity and the risk of death from prostate cancer after radical prostatectomy. N Engl J Med2004; 351:125-135
  87. Loeb S, Metter EJ, Kan D, Roehl KA, Catalona WJ. Prostate-specific antigen velocity (PSAV) risk count improves the specificity of screening for clinically significant prostate cancer. BJU international2012; 109:508-513; discussion 513-504
  88. Loeb S, Carter HB, Schaeffer EM, Kettermann A, Ferrucci L, Metter EJ. Distribution of PSA velocity by total PSA levels: data from the Baltimore Longitudinal Study of Aging. Urology2011; 77:143-147
  89. Clements JA, Merritt T, DeVoss K, Swanson C, Hamlyn L, Scells B, Rohde P, Lavin MF, Yaxley J, Gardiner RA. Inactive free : total prostate specific antigen ratios in ejaculate from men with suspected and known prostate cancer, compared with young control men. BJU international2000; 86:453-458
  90. Savblom C, Malm J, Giwercman A, Nilsson JA, Berglund G, Lilja H. Blood levels of free-PSA but not complex-PSA significantly correlates to prostate release of PSA in semen in young men, while blood levels of complex-PSA, but not free-PSA increase with age. The Prostate2005; 65:66-72
  91. Stenman UH, Abrahamsson PA, Aus G, Lilja H, Bangma C, Hamdy FC, Boccon-Gibod L, Ekman P. Prognostic value of serum markers for prostate cancer. Scandinavian journal of urology and nephrology Supplementum2005:64-81
  92. Lilja H. Significance of different molecular forms of serum PSA. The free, noncomplexed form of PSA versus that complexed to alpha 1-antichymotrypsin. The Urologic clinics of North America1993; 20:681-686
  93. Seaman E, Whang M, Olsson CA, Katz A, Cooner WH, Benson MC. PSA density (PSAD). Role in patient evaluation and management. The Urologic clinics of North America1993; 20:653-663
  94. Lughezzani G, Lazzeri M, Haese A, McNicholas T, de la Taille A, Buffi NM, Fossati N, Lista G, Larcher A, Abrate A, Mistretta A, Bini V, Redorta JP, Graefen M, Guazzoni G. Multicenter European External Validation of a Prostate Health Index-based Nomogram for Predicting Prostate Cancer at Extended Biopsy. Eur Urol2013;
  95. Fossati N, Buffi NM, Haese A, Stephan C, Larcher A, McNicholas T, de la Taille A, Freschi M, Lughezzani G, Abrate A, Bini V, Palou Redorta J, Graefen M, Guazzoni G, Lazzeri M. Preoperative Prostate-specific Antigen Isoform p2PSA and Its Derivatives, %p2PSA and Prostate Health Index, Predict Pathologic Outcomes in Patients Undergoing Radical Prostatectomy for Prostate Cancer: Results from a Multicentric European Prospective Study. European Urology2015; 68:132-138
  96. Hori S, Blanchet JS, McLoughlin J. From prostate-specific antigen (PSA) to precursor PSA (proPSA) isoforms: a review of the emerging role of proPSAs in the detection and management of early prostate cancer. BJU international2013; 112:717-728
  97. Catalona WJ, Partin AW, Sanda MG, Wei JT, Klee GG, Bangma CH, Slawin KM, Marks LS, Loeb S, Broyles DL, Shin SS, Cruz AB, Chan DW, Sokoll LJ, Roberts WL, van Schaik RHN, Mizrahi IA. A Multicenter Study of [-2]Pro-Prostate Specific Antigen Combined With Prostate Specific Antigen and Free Prostate Specific Antigen for Prostate Cancer Detection in the 2.0 to 10.0 ng/ml Prostate Specific Antigen Range. The Journal of urology2011; 185:1650-1655
  98. Ferro M, Bruzzese D, Perdonà S, Marino A, Mazzarella C, Perruolo G, D’Esposito V, Cosimato V, Buonerba C, Di Lorenzo G, Musi G, De Cobelli O, Chun FK, Terracciano D. Prostate Health Index (Phi) and Prostate Cancer Antigen 3 (PCA3) Significantly Improve Prostate Cancer Detection at Initial Biopsy in a Total PSA Range of 2–10 ng/ml. PLOS ONE2013; 8:e67687
  99. Lazzeri M, Haese A, de la Taille A, Palou Redorta J, McNicholas T, Lughezzani G, Scattoni V, Bini V, Freschi M, Sussman A, Ghaleh B, Le Corvoisier P, Alberola Bou J, Esquena Fernández S, Graefen M, Guazzoni G. Serum Isoform [−2]proPSA Derivatives Significantly Improve Prediction of Prostate Cancer at Initial Biopsy in a Total PSA Range of 2–10 ng/ml: A Multicentric European Study. European Urology2013; 63:986-994
  100. Loeb S, Sanda MG, Broyles DL, Shin SS, Bangma CH, Wei JT, Partin AW, Klee GG, Slawin KM, Marks LS, van Schaik RHN, Chan DW, Sokoll LJ, Cruz AB, Mizrahi IA, Catalona WJ. The Prostate Health Index Selectively Identifies Clinically Significant Prostate Cancer. The Journal of urology2015; 193:1163-1169
  101. de la Calle C, Patil D, Wei JT, Scherr DS, Sokoll L, Chan DW, Siddiqui J, Mosquera JM, Rubin MA, Sanda MG. Multicenter Evaluation of the Prostate Health Index to Detect Aggressive Prostate Cancer in Biopsy Naïve Men. The Journal of urology2015; 194:65-72
  102. Filella X, Giménez N. Evaluation of [−2] proPSA and Prostate Health Index (phi) for the detection of prostate cancer: a systematic review and meta-analysis. Clinical Chemistry and Laboratory Medicine.Vol 512013:729.
  103. Lughezzani G, Lazzeri M, Larcher A, Lista G, Scattoni V, Cestari A, Buffi NM, Bini V, Guazzoni G. Development and internal validation of a Prostate Health Index based nomogram for predicting prostate cancer at extended biopsy. The Journal of urology2012; 188:1144-1150
  104. Druskin SC, Tosoian JJ, Young A, Collica S, Srivastava A, Ghabili K, Macura KJ, Carter BH, Partin AW, Sokoll LJ, Ross AE, Pavlovich CP. Incorporating Prostate Health Index Density, MRI, and Prior Negative Biopsy Status to Improve the Detection of Clinically Significant Prostate Cancer. BJU international2017;
  105. Nordström T, Vickers A, Assel M, Lilja H, Grönberg H, Eklund M. Comparison Between the Four-kallikrein Panel and Prostate Health Index for Predicting Prostate Cancer. European Urology2015; 68:139-146
  106. Braun K, Sjoberg DD, Vickers AJ, Lilja H, Bjartell AS. A Four-kallikrein Panel Predicts High-grade Cancer on Biopsy: Independent Validation in a Community Cohort. European Urology2016; 69:505-511
  107. Parekh DJ, Punnen S, Sjoberg DD, Asroff SW, Bailen JL, Cochran JS, Concepcion R, David RD, Deck KB, Dumbadze I, Gambla M, Grable MS, Henderson RJ, Karsh L, Krisch EB, Langford TD, Lin DW, McGee SM, Munoz JJ, Pieczonka CM, Rieger-Christ K, Saltzstein DR, Scott JW, Shore ND, Sieber PR, Waldmann TM, Wolk FN, Zappala SM. A multi-institutional prospective trial in the USA confirms that the 4Kscore accurately identifies men with high-grade prostate cancer. Eur Urol2015; 68:464-470
  108. Bryant RJ, Sjoberg DD, Vickers AJ, Robinson MC, Kumar R, Marsden L, Davis M, Scardino PT, Donovan J, Neal DE, Lilja H, Hamdy FC. Predicting High-Grade Cancer at Ten-Core Prostate Biopsy Using Four Kallikrein Markers Measured in Blood in the ProtecT Study. JNCI: Journal of the National Cancer Institute2015; 107:djv095-djv095
  109. Brawer MK. The diagnosis of prostatic carcinoma. Cancer1993; 71:899-905
  110. Jewett HJ. Significance of the palpable prostatic nodule. Journal of the American Medical Association1956; 160:838-839
  111. Catalona WJ, Richie JP, Ahmann FR, Hudson MA, Scardino PT, Flanigan RC, deKernion JB, Ratliff TL, Kavoussi LR, Dalkin BL, et al. Comparison of digital rectal examination and serum prostate specific antigen in the early detection of prostate cancer: results of a multicenter clinical trial of 6,630 men. The Journal of urology1994; 151:1283-1290
  112. Ellis WJ, Chetner MP, Preston SD, Brawer MK. Diagnosis of prostatic carcinoma: the yield of serum prostate specific antigen, digital rectal examination and transrectal ultrasonography. The Journal of urology1994; 152:1520-1525
  113. Prostate-specific antigen (PSA) best practice policy. American Urological Association (AUA). Oncology (Williston Park, NY)2000; 14:267-272, 277-268, 280 passim
  114. Partin AW, Yoo J, Carter HB, Pearson JD, Chan DW, Epstein JI, Walsh PC. The use of prostate specific antigen, clinical stage and Gleason score to predict pathological stage in men with localized prostate cancer. The Journal of urology1993; 150:110-114
  115. Thompson IM, Rounder JB, Teague JL, Peek M, Spence CR. Impact of routine screening for adenocarcinoma of the prostate on stage distribution. The Journal of urology1987; 137:424-426
  116. McLaughlin AP, Saltzstein SL, McCullough DL, Gittes RF. Prostatic carcinoma: incidence and location of unsuspected lymphatic metastases. The Journal of urology1976; 115:89-94
  117. Bussemakers MJ, van BA, Verhaegh GW, Smit FP, Karthaus HF, Schalken JA, Debruyne FM, Ru N, Isaacs WB. DD3: a new prostate-specific gene, highly overexpressed in prostate cancer. Cancer Res1999; 59:5975-5979
  118. Fradet Y, Saad F, Aprikian A, Dessureault J, Elhilali M, Trudel C, Masse B, Piche L, Chypre C. uPM3, a new molecular urine test for the detection of prostate cancer. Urology2004; 64:311-315; discussion 315-316
  119. Landers KA, Burger MJ, Tebay MA, Purdie DM, Scells B, Samaratunga H, Lavin MF, Gardiner RA. Use of multiple biomarkers for a molecular diagnosis of prostate cancer. International Journal of Cancer2005; 114:950-956
  120. Hessels D, Klein GJM, van Oort I, Karthaus HF, van Leenders GJL, van Balken B, Kiemeney LA, Witjes JA, Schalken JA. DD3(PCA3)-based molecular urine analysis for the diagnosis of prostate cancer. European Urology2003; 44:8-15; discussion 15-16
  121. Marks LS, Fradet Y, Deras IL, Blase A, Mathis J, Aubin SM, Cancio AT, Desaulniers M, Ellis WJ, Rittenhouse H, Groskopf J. PCA3 molecular urine assay for prostate cancer in men undergoing repeat biopsy. Urology2007; 69:532-535
  122. Roobol MJ, Schroder FH, van Leeuwen P, Wolters T, van den Bergh RC, van Leenders GJ, Hessels D. Performance of the prostate cancer antigen 3 (PCA3) gene and prostate-specific antigen in prescreened men: exploring the value of PCA3 for a first-line diagnostic test. Eur Urol2010; 58:475-481
  123. Nyberg M, Ulmert D, Lindgren A, Lindstrom U, Abrahamsson PA, Bjartell A. PCA3 as a diagnostic marker for prostate cancer: a validation study on a Swedish patient population. Scandinavian journal of urology and nephrology2010; 44:378-383
  124. Roobol MJ. Contemporary role of prostate cancer gene 3 in the management of prostate cancer. Current opinion in urology2011; 21:225-229
  125. Wei JT, Feng Z, Partin AW, Brown E, Thompson I, Sokoll L, Chan DW, Lotan Y, Kibel AS, Busby JE, Bidair M, Lin DW, Taneja SS, Viterbo R, Joon AY, Dahlgren J, Kagan J, Srivastava S, Sanda MG. Can Urinary PCA3 Supplement PSA in the Early Detection of Prostate Cancer? Journal of clinical oncology : official journal of the American Society of Clinical Oncology2014; 32:4066-4072
  126. Tinzl M, Marberger M, Horvath S, Chypre C. DD3PCA3 RNA analysis in urine--a new perspective for detecting prostate cancer. Eur Urol2004; 46:182-186; discussion 187
  127. van Gils MP, Hessels D, van Hooij O, Jannink SA, Peelen WP, Hanssen SL, Witjes JA, Cornel EB, Karthaus HF, Smits GA, Dijkman GA, Mulders PF, Schalken JA. The time-resolved fluorescence-based PCA3 test on urinary sediments after digital rectal examination; a Dutch multicenter validation of the diagnostic performance. Clinical cancer research : an official journal of the American Association for Cancer Research2007; 13:939-943
  128. van Gils MP, Cornel EB, Hessels D, Peelen WP, Witjes JA, Mulders PF, Rittenhouse HG, Schalken JA. Molecular PCA3 diagnostics on prostatic fluid. The Prostate2007; 67:881-887
  129. Salami SS, Schmidt F, Laxman B, Regan MM, Rickman DS, Scherr D, Bueti G, Siddiqui J, Tomlins SA, Wei JT, Chinnaiyan AM, Rubin MA, Sanda MG. Combining urinary detection of TMPRSS2:ERG and PCA3 with serum PSA to predict diagnosis of prostate cancer. Urol Oncol2013; 31:566-571
  130. Roberts MJ, Richards RS, Gardiner RA, Selth LA. Seminal fluid: a useful source of prostate cancer biomarkers? Biomarkers in medicine2015; 9:77-80
  131. Roberts MJ, Richards RS, Chow CW, Doi SA, Schirra HJ, Buck M, Samaratunga H, Perry-Keene J, Payton D, Yaxley J, Lavin MF, Gardiner RA. Prostate-based biofluids for the detection of prostate cancer: A comparative study of the diagnostic performance of cell-sourced RNA biomarkers. Prostate international2016; 4:97-102
  132. Roberts MJ, Chow CW, Schirra HJ, Richards R, Buck M, Selth LA, Doi SA, Samaratunga H, Perry-Keene J, Payton D, Yaxley J, Lavin MF, Gardiner RA. Diagnostic performance of expression of PCA3, Hepsin and miR biomarkers inejaculate in combination with serum PSA for the detection of prostate cancer. The Prostate2015; 75:539-549
  133. Leyten GH, Hessels D, Jannink SA, Smit FP, de Jong H, Cornel EB, de Reijke TM, Vergunst H, Kil P, Knipscheer BC, van Oort IM, Mulders PF, Hulsbergen-van de Kaa CA, Schalken JA. Prospective multicentre evaluation of PCA3 and TMPRSS2-ERG gene fusions as diagnostic and prognostic urinary biomarkers for prostate cancer. Eur Urol2014; 65:534-542
  134. Tomlins SA, Aubin SM, Siddiqui J, Lonigro RJ, Sefton-Miller L, Miick S, Williamsen S, Hodge P, Meinke J, Blase A, Penabella Y, Day JR, Varambally R, Han B, Wood D, Wang L, Sanda MG, Rubin MA, Rhodes DR, Hollenbeck B, Sakamoto K, Silberstein JL, Fradet Y, Amberson JB, Meyers S, Palanisamy N, Rittenhouse H, Wei JT, Groskopf J, Chinnaiyan AM. Urine TMPRSS2:ERG fusion transcript stratifies prostate cancer risk in men with elevated serum PSA. Sci Transl Med2011; 3:94ra72
  135. Stephan C, Jung K, Semjonow A, Schulze-Forster K, Cammann H, Hu X, Meyer HA, Bogemann M, Miller K, Friedersdorff F. Comparative assessment of urinary prostate cancer antigen 3 and TMPRSS2:ERG gene fusion with the serum [-2]proprostate-specific antigen-based prostate health index for detection of prostate cancer. Clinical chemistry2013; 59:280-288
  136. Sanda MG, Feng Z, Howard DH, Tomlins SA, Sokoll LJ, Chan DW, Regan MM, Groskopf J, Chipman J, Patil DH, Salami SS, Scherr DS, Kagan J, Srivastava S, Thompson IM, Jr., Siddiqui J, Fan J, Joon AY, Bantis LE, Rubin MA, Chinnayian AM, Wei JT, Bidair M, Kibel A, Lin DW, Lotan Y, Partin A, Taneja S. Association Between Combined TMPRSS2:ERG and PCA3 RNA Urinary Testing and Detection of Aggressive Prostate Cancer. JAMA oncology2017; 3:1085-1093
  137. Gardiner RA, Mainwaring P, Lavin MF. Integrating Molecular Profiling of Liquid Biopsy Samples with a Calculator Algorithm To Detect High-risk Prostate Cancer. Eur Urol2016; 70:749-750
  138. Dickinson L, Ahmed HU, Allen C, Barentsz JO, Carey B, Futterer JJ, Heijmink SW, Hoskin PJ, Kirkham A, Padhani AR, Persad R, Puech P, Punwani S, Sohaib AS, Tombal B, Villers A, van der Meulen J, Emberton M. Magnetic resonance imaging for the detection, localisation, and characterisation of prostate cancer: recommendations from a European consensus meeting. Eur Urol2011; 59:477-494
  139. Pokorny MR, de Rooij M, Duncan E, Schroder FH, Parkinson R, Barentsz JO, Thompson LC. Prospective study of diagnostic accuracy comparing prostate cancer detection by transrectal ultrasound-guided biopsy versus magnetic resonance (MR) imaging with subsequent MR-guided biopsy in men without previous prostate biopsies. Eur Urol2014; 66:22-29
  140. Thompson J, Lawrentschuk N, Frydenberg M, Thompson L, Stricker P, Usanz. The role of magnetic resonance imaging in the diagnosis and management of prostate cancer. BJU international2013; 112 Suppl 2:6-20
  141. Thompson JE, Moses D, Shnier R, Brenner P, Delprado W, Ponsky L, Pulbrook M, Bohm M, Haynes AM, Hayen A, Stricker PD. Multiparametric magnetic resonance imaging guided diagnostic biopsy detects significant prostate cancer and could reduce unnecessary biopsies and over detection: a prospective study. The Journal of urology2014; 192:67-74
  142. Barentsz JO, Weinreb JC, Verma S, Thoeny HC, Tempany CM, Shtern F, Padhani AR, Margolis D, Macura KJ, Haider MA, Cornud F, Choyke PL. Synopsis of the PI-RADS v2 Guidelines for Multiparametric Prostate Magnetic Resonance Imaging and Recommendations for Use. Eur Urol2016; 69:41-49
  143. Moore CM, Kasivisvanathan V, Eggener S, Emberton M, Futterer JJ, Gill IS, Grubb Iii RL, Hadaschik B, Klotz L, Margolis DJ, Marks LS, Melamed J, Oto A, Palmer SL, Pinto P, Puech P, Punwani S, Rosenkrantz AB, Schoots IG, Simon R, Taneja SS, Turkbey B, Ukimura O, van der Meulen J, Villers A, Watanabe Y, Consortium S. Standards of reporting for MRI-targeted biopsy studies (START) of the prostate: recommendations from an International Working Group. Eur Urol2013; 64:544-552
  144. Rosenkrantz AB, Taneja SS. Radiologist, be aware: ten pitfalls that confound the interpretation of multiparametric prostate MRI. AJR Am J Roentgenol2014; 202:109-120
  145. Kuru TH, Roethke MC, Rieker P, Roth W, Fenchel M, Hohenfellner M, Schlemmer HP, Hadaschik BA. Histology core-specific evaluation of the European Society of Urogenital Radiology (ESUR) standardised scoring system of multiparametric magnetic resonance imaging (mpMRI) of the prostate. BJU international2013; 112:1080-1087
  146. de Rooij M, Hamoen EH, Futterer JJ, Barentsz JO, Rovers MM. Accuracy of multiparametric MRI for prostate cancer detection: a meta-analysis. AJR Am J Roentgenol2014; 202:343-351
  147. Futterer JJ. Multiparametric MRI in the Detection of Clinically Significant Prostate Cancer. Korean J Radiol2017; 18:597-606
  148. Ahmed HU, El-Shater Bosaily A, Brown LC, Gabe R, Kaplan R, Parmar MK, Collaco-Moraes Y, Ward K, Hindley RG, Freeman A, Kirkham AP, Oldroyd R, Parker C, Emberton M, group Ps. Diagnostic accuracy of multi-parametric MRI and TRUS biopsy in prostate cancer (PROMIS): a paired validating confirmatory study. Lancet (London, England)2017; 389:815-822
  149. Woo S, Suh CH, Kim SY, Cho JY, Kim SH. Diagnostic Performance of Prostate Imaging Reporting and Data System Version 2 for Detection of Prostate Cancer: A Systematic Review and Diagnostic Meta-analysis. Eur Urol2017; 72:177-188
  150. Roehl KA, Antenor JA, Catalona WJ. Serial biopsy results in prostate cancer screening study. The Journal of urology2002; 167:2435-2439
  151. Shinohara K, Nguyen H, Masic S. Management of an increasing prostate-specific antigen level after negative prostate biopsy. The Urologic clinics of North America2014; 41:327-338
  152. Hambrock T, Somford DM, Hoeks C, Bouwense SA, Huisman H, Yakar D, van Oort IM, Witjes JA, Futterer JJ, Barentsz JO. Magnetic resonance imaging guided prostate biopsy in men with repeat negative biopsies and increased prostate specific antigen. The Journal of urology2010; 183:520-527
  153. Hoeks CM, Schouten MG, Bomers JG, Hoogendoorn SP, Hulsbergen-van de Kaa CA, Hambrock T, Vergunst H, Sedelaar JP, Futterer JJ, Barentsz JO. Three-Tesla magnetic resonance-guided prostate biopsy in men with increased prostate-specific antigen and repeated, negative, random, systematic, transrectal ultrasound biopsies: detection of clinically significant prostate cancers. Eur Urol2012; 62:902-909
  154. Truong M, Frye TP. Magnetic resonance imaging detection of prostate cancer in men with previous negative prostate biopsy. Transl Androl Urol2017; 6:424-431
  155. Daneshgari F, Taylor GD, Miller GJ, Crawford ED. Computer simulation of the probability of detecting low volume carcinoma of the prostate with six random systematic core biopsies. Urology1995; 45:604-609
  156. Gore JL, Shariat SF, Miles BJ, Kadmon D, Jiang N, Wheeler TM, Slawin KM. Optimal combinations of systematic sextant and laterally directed biopsies for the detection of prostate cancer. The Journal of urology2001; 165:1554-1559
  157. Presti JC, Jr., Chang JJ, Bhargava V, Shinohara K. The optimal systematic prostate biopsy scheme should include 8 rather than 6 biopsies: results of a prospective clinical trial. The Journal of urology2000; 163:163-166; discussion 166-167
  158. Vashi AR, Wojno KJ, Gillespie B, Oesterling JE. A model for the number of cores per prostate biopsy based on patient age and prostate gland volume. The Journal of urology1998; 159:920-924
  159. Siddiqui MM, Rais-Bahrami S, Turkbey B, George AK, Rothwax J, Shakir N, Okoro C, Raskolnikov D, Parnes HL, Linehan WM, Merino MJ, Simon RM, Choyke PL, Wood BJ, Pinto PA. Comparison of MR/ultrasound fusion-guided biopsy with ultrasound-guided biopsy for the diagnosis of prostate cancer. Jama2015; 313:390-397
  160. Schaeffer EM, Carter HB, Kettermann A, Loeb S, Ferrucci L, Landis P, Trock BJ, Metter EJ. Prostate specific antigen testing among the elderly--when to stop? The Journal of urology2009; 181:1606-1614; discussion 1613-1604
  161. Mottet N, Bellmunt J, Bolla M, Briers E, Cumberbatch MG, De Santis M, Fossati N, Gross T, Henry AM, Joniau S, Lam TB, Mason MD, Matveev VB, Moldovan PC, van den Bergh RCN, Van den Broeck T, van der Poel HG, van der Kwast TH, Rouviere O, Schoots IG, Wiegel T, Cornford P. EAU-ESTRO-SIOG Guidelines on Prostate Cancer. Part 1: Screening, Diagnosis, and Local Treatment with Curative Intent. Eur Urol2017; 71:618-629
  162. Loeb S, Carter HB, Berndt SI, Ricker W, Schaeffer EM. Complications after prostate biopsy: data from SEER-Medicare. The Journal of urology2011; 186:1830-1834
  163. Ismail MT, Gomella LG. Transrectal prostate biopsy. The Urologic clinics of North America2013; 40:457-472
  164. Loeb S, Vellekoop A, Ahmed HU, Catto J, Emberton M, Nam R, Rosario DJ, Scattoni V, Lotan Y. Systematic review of complications of prostate biopsy. Eur Urol2013; 64:876-892
  165. Zani EL, Clark OA, Rodrigues Netto N, Jr. Antibiotic prophylaxis for transrectal prostate biopsy. Cochrane Database Syst Rev2011:CD006576
  166. Liss MA, Ehdaie B, Loeb S, Meng MV, Raman JD, Spears V, Stroup SP. An Update of the American Urological Association White Paper on the Prevention and Treatment of the More Common Complications Related to Prostate Biopsy. The Journal of urology2017;
  167. Carignan A, Roussy JF, Lapointe V, Valiquette L, Sabbagh R, Pepin J. Increasing risk of infectious complications after transrectal ultrasound-guided prostate biopsies: time to reassess antimicrobial prophylaxis? Eur Urol2012; 62:453-459
  168. Borghesi M, Ahmed H, Nam R, Schaeffer E, Schiavina R, Taneja S, Weidner W, Loeb S. Complications After Systematic, Random, and Image-guided Prostate Biopsy. Eur Urol2017; 71:353-365
  169. Abughosh Z, Margolick J, Goldenberg SL, Taylor SA, Afshar K, Bell R, Lange D, Bowie WR, Roscoe D, Machan L, Black PC. A prospective randomized trial of povidone-iodine prophylactic cleansing of the rectum before transrectal ultrasound guided prostate biopsy. The Journal of urology2013; 189:1326-1331
  170. Bennett HY, Roberts MJ, Doi SA, Gardiner RA. The global burden of major infectious complications following prostate biopsy. Epidemiol Infect2016; 144:1784-1791
  171. Williamson DA, Roberts SA, Paterson DL, Sidjabat H, Silvey A, Masters J, Rice M, Freeman JT. Escherichia coli bloodstream infection after transrectal ultrasound-guided prostate biopsy: implications of fluoroquinolone-resistant sequence type 131 as a major causative pathogen. Clin Infect Dis2012; 54:1406-1412
  172. Roberts MJ, Williamson DA, Hadway P, Doi SA, Gardiner RA, Paterson DL. Baseline prevalence of antimicrobial resistance and subsequent infection following prostate biopsy using empirical or altered prophylaxis: A bias-adjusted meta-analysis. International journal of antimicrobial agents2014; 43:301-309
  173. Manecksha RP, Nason GJ, Cullen IM, Fennell JP, McEvoy E, McDermott T, Flynn RJ, Grainger R, Thornhill JA. Prospective study of antibiotic prophylaxis for prostate biopsy involving >1100 men. ScientificWorldJournal2012; 2012:650858
  174. Roberts MJ, Bennett HY, Harris PN, Holmes M, Grummet J, Naber K, Wagenlehner FME. Prostate Biopsy-related Infection: A Systematic Review of Risk Factors, Prevention Strategies, and Management Approaches. Urology2017; 104:11-21
  175. Zowawi HM, Harris PN, Roberts MJ, Tambyah PA, Schembri MA, Pezzani MD, Williamson DA, Paterson DL. The emerging threat of multidrug-resistant Gram-negative bacteria in urology. Nat Rev Urol2015; 12:570-584
  176. Yaxley AJ, Yaxley JW, Thangasamy IA, Ballard E, Pokorny MR. Comparison between target magnetic resonance imaging (MRI) in-gantry and cognitively directed transperineal or transrectal-guided prostate biopsies for Prostate Imaging-Reporting and Data System (PI-RADS) 3-5 MRI lesions. BJU international2017; 120 Suppl 3:43-50
  177. Bains LJ, Studer UE, Froehlich JM, Giannarini G, Triantafyllou M, Fleischmann A, Thoeny HC. Diffusion-weighted magnetic resonance imaging detects significant prostate cancer with high probability. The Journal of urology2014; 192:737-742
  178. Thompson JE, van Leeuwen PJ, Moses D, Shnier R, Brenner P, Delprado W, Pulbrook M, Bohm M, Haynes AM, Hayen A, Stricker PD. The Diagnostic Performance of Multiparametric Magnetic Resonance Imaging to Detect Significant Prostate Cancer. The Journal of urology2016; 195:1428-1435
  179. Epstein JI, Amin MB, Reuter VE, Humphrey PA. Contemporary Gleason Grading of Prostatic Carcinoma: An Update With Discussion on Practical Issues to Implement the 2014 International Society of Urological Pathology (ISUP) Consensus Conference on Gleason Grading of Prostatic Carcinoma. Am J Surg Pathol2017; 41:e1-e7
  180. Amin MB, Lin DW, Gore JL, Srigley JR, Samaratunga H, Egevad L, Rubin M, Nacey J, Carter HB, Klotz L, Sandler H, Zietman AL, Holden S, Montironi R, Humphrey PA, Evans AJ, Epstein JI, Delahunt B, McKenney JK, Berney D, Wheeler TM, Chinnaiyan AM, True L, Knudsen B, Hammond ME. The critical role of the pathologist in determining eligibility for active surveillance as a management option in patients with prostate cancer: consensus statement with recommendations supported by the College of American Pathologists, International Society of Urological Pathology, Association of Directors of Anatomic and Surgical Pathology, the New Zealand Society of Pathologists, and the Prostate Cancer Foundation. Archives of pathology & laboratory medicine2014; 138:1387-1405
  181. de la Taille A, Katz A, Bagiella E, Olsson CA, O'Toole KM, Rubin MA. Perineural invasion on prostate needle biopsy: an independent predictor of final pathologic stage. Urology1999; 54:1039-1043
  182. Sanwick JM, Dalkin BL, Nagle RB. Accuracy of prostate needle biopsy in predicting extracapsular tumor extension at radical retropubic prostatectomy: application in selecting patients for nerve-sparing surgery. Urology1998; 52:814-818; discussion 818-819
  183. Sebo TJ, Bock BJ, Cheville JC, Lohse C, Wollan P, Zincke H. The percent of cores positive for cancer in prostate needle biopsy specimens is strongly predictive of tumor stage and volume at radical prostatectomy. The Journal of urology2000; 163:174-178
  184. Egevad L, Delahunt B, Kristiansen G, Samaratunga H, Varma M. Contemporary prognostic indicators for prostate cancer incorporating International Society of Urological Pathology recommendations. Pathology2018; 50:60-73
  185. Srigley JR, Delahunt B, Egevad L, Samaratunga H, Yaxley J, Evans AJ. One is the new six: The International Society of Urological Pathology (ISUP) patient-focused approach to Gleason grading. Canadian Urological Association journal = Journal de l'Association des urologues du Canada2016; 10:339-341
  186. Epstein JI, Partin AW, Sauvageot J, Walsh PC. Prediction of progression following radical prostatectomy. A multivariate analysis of 721 men with long-term follow-up. Am J Surg Pathol1996; 20:286-292
  187. McNeal JE, Villers AA, Redwine EA, Freiha FS, Stamey TA. Histologic differentiation, cancer volume, and pelvic lymph node metastasis in adenocarcinoma of the prostate. Cancer1990; 66:1225-1233
  188. Sauter G, Steurer S, Clauditz TS, Krech T, Wittmer C, Lutz F, Lennartz M, Janssen T, Hakimi N, Simon R, von Petersdorff-Campen M, Jacobsen F, von Loga K, Wilczak W, Minner S, Tsourlakis MC, Chirico V, Haese A, Heinzer H, Beyer B, Graefen M, Michl U, Salomon G, Steuber T, Budaus LH, Hekeler E, Malsy-Mink J, Kutzera S, Fraune C, Gobel C, Huland H, Schlomm T. Clinical Utility of Quantitative Gleason Grading in Prostate Biopsies and Prostatectomy Specimens. Eur Urol2016; 69:592-598
  189. Egevad L, Delahunt B, Evans AJ, Grignon DJ, Kench JG, Kristiansen G, Leite KR, Samaratunga H, Srigley JR. International Society of Urological Pathology (ISUP) Grading of Prostate Cancer. Am J Surg Pathol2016; 40:858-861
  190. Samaratunga H, Delahunt B, Gianduzzo T, Coughlin G, Duffy D, LeFevre I, Johannsen S, Egevad L, Yaxley J. The prognostic significance of the 2014 International Society of Urological Pathology (ISUP) grading system for prostate cancer. Pathology2015; 47:515-519
  191. Delahunt B, Egevad L, Srigley JR, Steigler A, Murray JD, Atkinson C, Matthews J, Duchesne G, Spry NA, Christie D, Joseph D, Attia J, Denham JW. Validation of International Society of Urological Pathology (ISUP) grading for prostatic adenocarcinoma in thin core biopsies using TROG 03.04 'RADAR' trial clinical data. Pathology2015; 47:520-525
  192. Bostwick DG, Brawer MK. Prostatic intra-epithelial neoplasia and early invasion in prostate cancer. Cancer1987; 59:788-794
  193. Bostwick DG, Pacelli A, Lopez-Beltran A. Molecular biology of prostatic intraepithelial neoplasia. The Prostate1996; 29:117-134
  194. Haggman MJ, Macoska JA, Wojno KJ, Oesterling JE. The relationship between prostatic intraepithelial neoplasia and prostate cancer: critical issues. The Journal of urology1997; 158:12-22
  195. Schlesinger C, Bostwick DG, Iczkowski KA. High-grade prostatic intraepithelial neoplasia and atypical small acinar proliferation: predictive value for cancer in current practice. Am J Surg Pathol2005; 29:1201-1207
  196. Chan TY, Epstein JI. Follow-up of atypical prostate needle biopsies suspicious for cancer. Urology1999; 53:351-355
  197. Allen EA, Kahane H, Epstein JI. Repeat biopsy strategies for men with atypical diagnoses on initial prostate needle biopsy. Urology1998; 52:803-807
  198. Buyyounouski MK, Choyke PL, McKenney JK, Sartor O, Sandler HM, Amin MB, Kattan MW, Lin DW. Prostate cancer – major changes in the American Joint Committee on Cancer eighth edition cancer staging manual. CA Cancer J Clin2017; 67:245-253
  199. Bartsch G, Horninger W, Klocker H, Pelzer A, Bektic J, Oberaigner W, Schennach H, Schafer G, Frauscher F, Boniol M, Severi G, Robertson C, Boyle P, Tyrol Prostate Cancer Screening G. Tyrol Prostate Cancer Demonstration Project: early detection, treatment, outcome, incidence and mortality. BJU international2008; 101:809-816
  200. Andriole GL, Crawford ED, Grubb RL, 3rd, Buys SS, Chia D, Church TR, Fouad MN, Gelmann EP, Kvale PA, Reding DJ, Weissfeld JL, Yokochi LA, O'Brien B, Clapp JD, Rathmell JM, Riley TL, Hayes RB, Kramer BS, Izmirlian G, Miller AB, Pinsky PF, Prorok PC, Gohagan JK, Berg CD, Team PP. Mortality results from a randomized prostate-cancer screening trial. N Engl J Med2009; 360:1310-1319
  201. Andriole GL, Crawford ED, Grubb RL, 3rd, Buys SS, Chia D, Church TR, Fouad MN, Isaacs C, Kvale PA, Reding DJ, Weissfeld JL, Yokochi LA, O'Brien B, Ragard LR, Clapp JD, Rathmell JM, Riley TL, Hsing AW, Izmirlian G, Pinsky PF, Kramer BS, Miller AB, Gohagan JK, Prorok PC, Team PP. Prostate cancer screening in the randomized Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial: mortality results after 13 years of follow-up. Journal of the National Cancer Institute2012; 104:125-132
  202. Schroder FH, Hugosson J, Roobol MJ, Tammela TL, Ciatto S, Nelen V, Kwiatkowski M, Lujan M, Lilja H, Zappa M, Denis LJ, Recker F, Paez A, Maattanen L, Bangma CH, Aus G, Carlsson S, Villers A, Rebillard X, van der Kwast T, Kujala PM, Blijenberg BG, Stenman UH, Huber A, Taari K, Hakama M, Moss SM, de Koning HJ, Auvinen A, Investigators E. Prostate-cancer mortality at 11 years of follow-up. N Engl J Med2012; 366:981-990
  203. Hugosson J, Carlsson S, Aus G, Bergdahl S, Khatami A, Lodding P, Pihl CG, Stranne J, Holmberg E, Lilja H. Mortality results from the Goteborg randomised population-based prostate-cancer screening trial. Lancet Oncol2010; 11:725-732
  204. Sandblom G, Varenhorst E, Rosell J, Lofman O, Carlsson P. Randomised prostate cancer screening trial: 20 year follow-up. BMJ (Clinical research ed)2011; 342:d1539
  205. Kjellman A, Akre O, Norming U, Tornblom M, Gustafsson O. 15-year followup of a population based prostate cancer screening study. The Journal of urology2009; 181:1615-1621; discussion 1621
  206. Labrie F, Candas B, Cusan L, Gomez JL, Belanger A, Brousseau G, Chevrette E, Levesque J. Screening decreases prostate cancer mortality: 11-year follow-up of the 1988 Quebec prospective randomized controlled trial. The Prostate2004; 59:311-318
  207. Shoag JE, Mittal S, Hu JC. Reevaluating PSA Testing Rates in the PLCO Trial. New England Journal of Medicine2016; 374:1795-1796
  208. Hugosson J, Carlsson SV. The dilemmas of prostate cancer screening. Med J Aust2013; 198:528-529
  209. Arsov C, Becker N, Hadaschik BA, Hohenfellner M, Herkommer K, Gschwend JE, Imkamp F, Kuczyk MA, Antoch G, Kristiansen G, Siener R, Semjonow A, Hamdy FC, Lilja H, Vickers AJ, Schroder FH, Albers P. Prospective randomized evaluation of risk-adapted prostate-specific antigen screening in young men: the PROBASE trial. Eur Urol2013; 64:873-875
  210. Gardiner RF, Yaxley J, Baade PD. Integrating disparate snippets of information in an approach to PSA testing in Australia and New Zealand. BJU international2012; 110 Suppl 4:35-37
  211. Stricker PD, Frydenberg M, Kneebone A, Chopra S. Informed prostate cancer risk-adjusted testing: a new paradigm. BJU international2012; 110 Suppl 4:30-34
  212. Schroder FH, Hugosson J, Roobol MJ, Tammela TL, Zappa M, Nelen V, Kwiatkowski M, Lujan M, Maattanen L, Lilja H, Denis LJ, Recker F, Paez A, Bangma CH, Carlsson S, Puliti D, Villers A, Rebillard X, Hakama M, Stenman UH, Kujala P, Taari K, Aus G, Huber A, van der Kwast TH, van Schaik RH, de Koning HJ, Moss SM, Auvinen A, Investigators E. Screening and prostate cancer mortality: results of the European Randomised Study of Screening for Prostate Cancer (ERSPC) at 13 years of follow-up. Lancet (London, England)2014; 384:2027-2035
  213. Tsodikov A, Gulati R, Heijnsdijk EAM, Pinsky PF, Moss SM, Qiu S, de Carvalho TM, Hugosson J, Berg CD, Auvinen A, Andriole GL, Roobol MJ, Crawford ED, Nelen V, Kwiatkowski M, Zappa M, Lujan M, Villers A, Feuer EJ, de Koning HJ, Mariotto AB, Etzioni R. Reconciling the Effects of Screening on Prostate Cancer Mortality in the ERSPC and PLCO Trials. Ann Intern Med2017; 167:449-455
  214. Vickers AJ. Prostate Cancer Screening: Time to Question How to Optimize the Ratio of Benefits and Harms. Ann Intern Med2017; 167:509-510
  215. Carlsson S, Assel M, Sjoberg D, Ulmert D, Hugosson J, Lilja H, Vickers A. Influence of blood prostate specific antigen levels at age 60 on benefits and harms of prostate cancer screening: population based cohort study. BMJ (Clinical research ed)2014; 348:g2296
  216. Litwin MS, Greenfield S, Elkin EP, Lubeck DP, Broering JM, Kaplan SH. Assessment of prognosis with the total illness burden index for prostate cancer: aiding clinicians in treatment choice. Cancer2007; 109:1777-1783
  217. Briganti A, Spahn M, Joniau S, Gontero P, Bianchi M, Kneitz B, Chun FK, Sun M, Graefen M, Abdollah F, Marchioro G, Frohenberg D, Giona S, Frea B, Karakiewicz PI, Montorsi F, Van Poppel H, Jeffrey Karnes R, European Multicenter Prostate Cancer C, Translational Research G. Impact of age and comorbidities on long-term survival of patients with high-risk prostate cancer treated with radical prostatectomy: a multi-institutional competing-risks analysis. Eur Urol2013; 63:693-701
  218. Lee JY, Lee DH, Cho NH, Rha KH, Choi YD, Hong SJ, Yang SC, Cho KS. Impact of Charlson comorbidity index varies by age in patients with prostate cancer treated by radical prostatectomy: a competing risk regression analysis. Ann Surg Oncol2014; 21:677-683
  219. Froehner M, Hentschel C, Koch R, Litz RJ, Hakenberg OW, Wirth MP. Which comorbidity classification best fits elderly candidates for radical prostatectomy? Urol Oncol2013; 31:461-467
  220. Chew KK, Gibson N, Sanfilippo F, Stuckey B, Bremner A. Cardiovascular mortality in men with erectile dysfunction: increased risk but not inevitable. J Sex Med2011; 8:1761-1771
  221. Banks E, Joshy G, Abhayaratna WP, Kritharides L, Macdonald PS, Korda RJ, Chalmers JP. Erectile dysfunction severity as a risk marker for cardiovascular disease hospitalisation and all-cause mortality: a prospective cohort study. PLoS Med2013; 10:e1001372
  222. Bill-Axelson A, Holmberg L, Garmo H, Rider JR, Taari K, Busch C, Nordling S, Haggman M, Andersson SO, Spangberg A, Andren O, Palmgren J, Steineck G, Adami HO, Johansson JE. Radical prostatectomy or watchful waiting in early prostate cancer. N Engl J Med2014; 370:932-942
  223. Vickers A, Bennette C, Steineck G, Adami HO, Johansson JE, Bill-Axelson A, Palmgren J, Garmo H, Holmberg L. Individualized estimation of the benefit of radical prostatectomy from the Scandinavian Prostate Cancer Group randomized trial. Eur Urol2012; 62:204-209
  224. Crawford ED, Grubb R, 3rd, Black A, Andriole GL, Jr., Chen MH, Izmirlian G, Berg CD, D'Amico AV. Comorbidity and mortality results from a randomized prostate cancer screening trial. Journal of clinical oncology : official journal of the American Society of Clinical Oncology2011; 29:355-361
  225. National Health Data Dictionary: version16.2.Australian Institute of Health and Welfare (AIHW);2015.
  226. Moyer VA, Force USPST. Screening for prostate cancer: U.S. Preventive Services Task Force recommendation statement. Ann Intern Med2012; 157:120-134
  227. Chambers SK, Zajdlewicz L, Youlden DR, Holland JC, Dunn J. The validity of the distress thermometer in prostate cancer populations. Psychooncology2014; 23:195-203
  228. Bill-Axelson A, Garmo H, Lambe M, Bratt O, Adolfsson J, Nyberg U, Steineck G, Stattin P. Suicide risk in men with prostate-specific antigen-detected early prostate cancer: a nationwide population-based cohort study from PCBaSe Sweden. Eur Urol2010; 57:390-395
  229. Carlsson S, Sandin F, Fall K, Lambe M, Adolfsson J, Stattin P, Bill-Axelson A. Risk of suicide in men with low-risk prostate cancer. Eur J Cancer2013; 49:1588-1599
  230. Fall K, Fang F, Mucci LA, Ye W, Andren O, Johansson JE, Andersson SO, Sparen P, Klein G, Stampfer M, Adami HO, Valdimarsdottir U. Immediate risk for cardiovascular events and suicide following a prostate cancer diagnosis: prospective cohort study. PLoS Med2009; 6:e1000197
  231. Chambers SK DJ, Lazenby M, Clutton S, Newton RU, Cormie P, Lowe A, Sandoe D, Gardiner RA. . ProsCare: A psychological care model for men with prostate cancer Australia: Prostate Cancer Foundation of Australia and Griffith University.
  232. Chambers SK, Hyde MK, Smith DP, Hughes S, Yuill S, Egger S, O'Connell DL, Stein K, Frydenberg M, Wittert G, Dunn J. New Challenges in Psycho‐Oncology Research III: A systematic review of psychological interventions for prostate cancer survivors and their partners: clinical and research implications. Psycho-Oncology2017; 26:873-913
  233. Goh AC, Kowalkowski MA, Bailey DE, Jr., Kazer MW, Knight SJ, Latini DM. Perception of cancer and inconsistency in medical information are associated with decisional conflict: a pilot study of men with prostate cancer who undergo active surveillance. BJU international2012; 110:E50-56
  234. Steginga SK, Occhipinti S, Gardiner RA, Yaxley J, Heathcote P. Making decisions about treatment for localized prostate cancer. BJU international2002; 89:255-260
  235. Chambers SK, Pinnock C, Lepore SJ, Hughes S, O'Connell DL. A systematic review of psychosocial interventions for men with prostate cancer and their partners. Patient Educ Couns2011; 85:e75-88
  236. Bill-Axelson A, Holmberg L, Ruutu M, Garmo H, Stark JR, Busch C, Nordling S, Haggman M, Andersson SO, Bratell S, Spangberg A, Palmgren J, Steineck G, Adami HO, Johansson JE, Investigators S-. Radical prostatectomy versus watchful waiting in early prostate cancer. N Engl J Med2011; 364:1708-1717
  237. Wilt TJ, Brawer MK, Jones KM, Barry MJ, Aronson WJ, Fox S, Gingrich JR, Wei JT, Gilhooly P, Grob BM, Nsouli I, Iyer P, Cartagena R, Snider G, Roehrborn C, Sharifi R, Blank W, Pandya P, Andriole GL, Culkin D, Wheeler T, Prostate Cancer Intervention versus Observation Trial Study G. Radical prostatectomy versus observation for localized prostate cancer. N Engl J Med2012; 367:203-213
  238. Barry MJ, Andriole GL, Culkin DJ, Fox SH, Jones KM, Carlyle MH, Wilt TJ. Ascertaining cause of death among men in the prostate cancer intervention versus observation trial. Clin Trials2013; 10:907-914
  239. Thompson IM, Jr., Tangen CM. Prostate cancer--uncertainty and a way forward. N Engl J Med2012; 367:270-271
  240. Clarke NW. The management of hormone-relapsed prostate cancer. BJU international2003; 92:860-868
  241. Melia J, Hewitson P, Austoker J. Introduction: Review of screening for prostate cancer. BJU international2005; 95 Suppl 3:1-3
  242. Frosch DL, Kaplan RM. Shared decision making in clinical medicine: past research and future directions. Am J Prev Med1999; 17:285-294
  243. Elwyn G, Edwards A, Kinnersley P. Shared decision-making in primary care: the neglected second half of the consultation. Br J Gen Pract1999; 49:477-482
  244. Davison BJ, Degner LF, Morgan TR. Information and decision-making preferences of men with prostate cancer. Oncol Nurs Forum1995; 22:1401-1408
  245. Davison BJ, Gleave ME, Goldenberg SL, Degner LF, Hoffart D, Berkowitz J. Assessing information and decision preferences of men with prostate cancer and their partners. Cancer Nurs2002; 25:42-49
  246. Barry MJ. Involving patients in medical decisions: how can physicians do better? Jama1999; 282:2356-2357
  247. Braddock CH, 3rd, Edwards KA, Hasenberg NM, Laidley TL, Levinson W. Informed decision making in outpatient practice: time to get back to basics. Jama1999; 282:2313-2320
  248. O'Connor AM, Bennett CL, Stacey D, Barry M, Col NF, Eden KB, Entwistle VA, Fiset V, Holmes-Rovner M, Khangura S, Llewellyn-Thomas H, Rovner D. Decision aids for people facing health treatment or screening decisions. Cochrane Database Syst Rev2009:CD001431
  249. Wilkinson S, Chodak G. Informed consent for prostate-specific antigen screening. Urology2003; 61:2-4
  250. Bird S. Risk management. Discussing benefits and risks with patients. PSA testing. Aust Fam Physician2004; 33:266-267
  251. Dunn IB, Kirk D. Legal pitfalls in the diagnosis of prostate cancer. BJU international2000; 86:304-307
  252. Carter HB. Informed consent for prostate-specific antigen screening. Urology2003; 61:13-14
  253. Ito K, Schroder FH. Informed consent for prostate-specific antigen-based screening - European view. Urology2003; 61:20-22
  254. Hewitson P, Austoker J. Part 2: Patient information, informed decision-making and the psycho-social impact of prostate-specific antigen testing. BJU international2005; 95 Suppl 3:16-32
  255. Chan EC, Sulmasy DP. What should men know about prostate-specific antigen screening before giving informed consent? Am J Med1998; 105:266-274
  256. Council GM. Consent: patients and doctors making decisions together.General Medical Council;2008.
  257. Bratt O, Damber JE, Emanuelsson M, Kristoffersson U, Lundgren R, Olsson H, Gronberg H. Risk perception, screening practice and interest in genetic testing among unaffected men in families with hereditary prostate cancer. Eur J Cancer2000; 36:235-241
  258. Brown CT, O'Flynn E, Van Der Meulen J, Newman S, Mundy AR, Emberton M. The fear of prostate cancer in men with lower urinary tract symptoms: should symptomatic men be screened? BJU international2003; 91:30-32
  259. Roberts RO, Rhodes T, Panser LA, Girman CJ, Chute CG, Oesterling JE, Lieber MM, Jacobsen SJ. Natural history of prostatism: worry and embarrassment from urinary symptoms and health care-seeking behavior. Urology1994; 43:621-628
  260. Lloyd A, Hayes P, Bell PR, Naylor AR. The role of risk and benefit perception in informed consent for surgery. Med Decis Making2001; 21:141-149
  261. Redelmeier DA, Rozin P, Kahneman D. Understanding patients' decisions. Cognitive and emotional perspectives. Jama1993; 270:72-76
  262. Baade PD, Steginga SK, Pinnock CB, Aitken JF. Communicating prostate cancer risk: what should we be telling our patients? Med J Aust2005; 182:472-475
  263. Gigerenzer G, Edwards A. Simple tools for understanding risks: from innumeracy to insight. BMJ (Clinical research ed)2003; 327:741-744
  264. Gurmankin AD, Baron J, Armstrong K. The effect of numerical statements of risk on trust and comfort with hypothetical physician risk communication. Med Decis Making2004; 24:265-271
  265. Paling J. Strategies to help patients understand risks. BMJ (Clinical research ed)2003; 327:745-748
  266. O'Connor AM, Rostom A, Fiset V, Tetroe J, Entwistle V, Llewellyn-Thomas H, Holmes-Rovner M, Barry M, Jones J. Decision aids for patients facing health treatment or screening decisions: systematic review. BMJ (Clinical research ed)1999; 319:731-734
  267. Chan EC, McFall SL, Byrd TL, Mullen PD, Volk RJ, Ureda J, Calderon-Mora J, Morales P, Valdes A, Kay Bartholomew L. A community-based intervention to promote informed decision making for prostate cancer screening among Hispanic American men changed knowledge and role preferences: a cluster RCT. Patient Educ Couns2011; 84:e44-51
  268. Lepore SJ, Wolf RL, Basch CE, Godfrey M, McGinty E, Shmukler C, Ullman R, Thomas N, Weinrich S. Informed decision making about prostate cancer testing in predominantly immigrant black men: a randomized controlled trial. Ann Behav Med2012; 44:320-330
  269. Myers RE, Daskalakis C, Kunkel EJ, Cocroft JR, Riggio JM, Capkin M, Braddock CH, 3rd. Mediated decision support in prostate cancer screening: a randomized controlled trial of decision counseling. Patient Educ Couns2011; 83:240-246
  270. Taylor KL, Williams RM, Davis K, Luta G, Penek S, Barry S, Kelly S, Tomko C, Schwartz M, Krist AH, Woolf SH, Fishman MB, Cole C, Miller E. Decision making in prostate cancer screening using decision aids vs usual care: a randomized clinical trial. JAMA Intern Med2013; 173:1704-1712

Hyponatremia

CLINICAL RECOGNITION

 

Hyponatremia is common, being found in some 15–20% of non-selected emergency admissions to hospitals. It is associated with increased mortality, morbidity and increased duration of hospital stay, independent of the cause of admission. Clinical presentation is diverse, ranging from seizure and coma at one extreme to apparent absence of symptoms. This spectrum reflects a number of influences (Figure 1).

 

- The rate of development of hyponatremia

- The degree of hyponatremia

- The neurophysiologic adaptive capacity of the individual

- The influence of co-morbidities


The management strategy is stratified, based on an overall assessment of the severity of the clinical presentation.

Plasma sodium concentration reflects the balance between sodium and water content, with each component reflecting the balance of intake and output. This approach can be used to develop a framework for classifying hyponatremia by etiology (Table 1).

Intravascular Volume Depletion (Hypovolemia)

 

Intravascular volume depletion leads to non-osmoregulated vasopressin (AVP) production and reduced free water excretion. AVP production can persist despite ensuing hyponatremia. Portal hypertension, congestive cardiac failure and hypoalbuminemia can all reduce effective circulating volume (independent of drug treatment), even in the context of excess total body sodium. Long-term diuretic use is a common cause of hyponatremia. Because it may develop slowly, patients can be relatively asymptomatic. Those on thiazide diuretics are particularly at risk as these agents produce solute loss without limiting renal concentrating ability.

 

Excess Hypotonic Fluid Intake

 

The administration or absorption of hypotonic fluids at a rate that exceeds renal free water excretion will inevitably result in hyponatremia. This can be seen with oral fluid intake (dipsogenic diabetes insipidus), intravenous fluid therapy and the absorption of hypotonic irrigating fluids following surgery to the lower renal tract or colonoscopy.

 

Syndrome of Inappropriate Antidiuresis (SIAD)

 

In SIAD there is a failure to maximally suppress AVP secretion as plasma osmolality falls below the normal osmotic threshold for AVP release. As patients continue to drink, persistent antidiuresis produces dilutional hyponatremia. Diagnosis of SIAD involves the exclusion of volume depletion and other endocrine causes of reduced free water excretion (Table 2).

 

Table 2. Diagnostic Criteria for SIAD

Hyponatremia
Urine Osmolality >100 mOsm/kg (sub-maximum dilution)
Urine Na+ >20 mmol/L (excluding effective intravascular volume depletion)
Absence of the following:

- hypotension and hypovolemia

- non-osmotic stimuli for AVP release

- oedema

- adrenal failure

- hypothyroidism

 

The majority of patients with SIAD are euvolemic on clinical examination. Urine sodium concentration is often above 80 mmol/l.

 

Many drugs cause SIAD. Drug histories are therefore an important part of the clinical assessment of patients presenting with hyponatremia (Table 3).

 

Table 3. Causes of SIAD

Drugs -  Antidepressants (Tricyclics, SSRIs)

-  Dopamine agonists (Metoclopramide, Prochlorperazine, Antipsychotics)

-  Anticonvulsants (Carbamazepine, Phenytoin, Sodium valproate)

-  Opiates

CNS Disturbances -  Stroke

-  Haemorrhage

-  Infection

-  Trauma

Malignancies -  Small cell lung cancer

-  Pancreatic, duodenal and head/neck cancers

Surgery -  Abdominal, thoracic or pituitary surgery
Pulmonary disease -  Pneumonia

-  Pneumothorax

Idiopathic

 

Central Salt Wasting

 

This acquired primary natriuresis is a very rare cause of hyponatremia with hypovolemia. The underlying mechanism(s) remains unclear but may involve a number of processes.

 

-  Increased release of natriuretic peptides

-  Reduced sympathetic drive

 

Central salt wasting has been described following a variety of neurosurgical situations. Diagnosis hinges on the natural history of the process.

 

-  The development of hyponatremia, preceded by natriuresis and diuresis

-  Clinical and biochemical features of hypovolemia and renal impairment

 

Central salt wasting is a concern for the neurosurgical patient in whom autoregulation of cerebral blood flow is disturbed such that small reductions in circulating volume can reduce cerebral perfusion. The syndrome of inappropriate antidiuresis (SIAD) can occur in the same group of patients. As management of the two conditions is diametrically opposed, it is important to make the correct diagnosis. Recent data suggest that salt wasting is indeed a very rare cause of hyponatremia in the neurosurgical patient.

 

Nephrogenic SIAD

 

The action of AVP on renal water excretion is mediated by the G-protein-coupled type 2 AVP-receptor (V2-R). Loss-of-function mutations of the V2-R are the cause of X-linked nephrogenic diabetes insipidus. The reciprocal phenotype, mutations in the V2-R that are constitutively activating, lead to AVP-independent but V2-R-mediated antidiuresis with persistent hyponatremia.  Patients may present in infancy or may remain undetected until adulthood.

 

DIAGNOSIS AND DIFFERENTIAL

 

History and examination are key to the clinical approach. They provide insights into the etiology, rate of development, clinical impact, and contributing co-morbidities: all factors important in a patient-focused approach to management (Table 4).

 

Table 4. Focus of History & Examination in the Patient with Hyponatremia

History -  Fluid loss (e.g. vomiting, diarrhoea)

-  Causes of SIAD

-  Symptoms of endocrine dysfunction suggestive of hypoadrenalism or hypopituitarism

-  Medication/drug use

Examination -  Signs of extracellular volume depletion

-  Orthostatic or persistent hypotension.

-  Signs of peripheral oedema, or ascites, heart failure, cirrhosis, or renal failure

 

Laboratory tests provide important information in the differential diagnosis of hyponatremia.

 

Serum Osmolality

 

The serum osmolality (which normally ranges from 275 to 290 mOsmol/kg) is primarily determined by the concentration of serum sodium (and accompanying anions). It is reduced in most cases of hyponatremia (hypotonic hyponatremia). In some instances, hyponatremia is not associated with dilute plasma. This is termed non-hypotonic hyponatremia (Table 5).

 

Urine Osmolality

 

The normal response to hyponatremia is to suppress AVP secretion, resulting in the excretion of maximally dilute urine with an osmolality below 100 mOsmol/kg. Values above this level indicate an inability to normally excrete free water, indicating secretion and action of AVP.

 

Urine Sodium Concentration

 

The urine sodium concentration can be used to distinguish between hyponatremia caused by effective arterial volume depletion (such as in true hypovolemia, heart failure, and cirrhosis) and that caused by euvolemic/hypervolemic hyponatremia (most often due to SIAD).

The combination of urine osmolality and urine Na+ concentration can be used to form a utilitarian algorithm for the investigation and differential diagnosis of hyponatremia (Figure 2).

TREATMENT

 

While hyponatremia can be life threatening, chronic hyponatremia can be tolerated very well even when profound. This diversity poses a management challenge: clinicians must balance the efficacy of any intervention with that of the potential adverse impact of both the intervention and persisting hyponatremia. Over-rapid correction of hyponatremia can trigger central nervous system osmotic demyelination. If urgent intervention is required, the aim of management should not be to normalise plasma sodium. Rather, it should be achieving a plasma sodium level that reverses or reduces morbidity while minimising the risk of osmotic demyelination. This requires a stratified approach based on clinical presentation: balancing a number of clinical drivers to optimise outcome (Figure 3).

General and Supportive Measures

 

The appropriate clinical environment for the management of hyponatremia is the one that matches the clinical needs of the patient. Management of hyponatremia in a patient with significant neurological morbidity, in whom plasma sodium is being raised over hours, requires close clinical and biochemical observation. This is best achieved in a high-dependency setting. Cerebral oedema and coma associated with hyponatremia may need supportive management with assisted ventilation.

 

Patients with Significant Coma or Seizures

 

Treatment with hypertonic sodium chloride increases plasma sodium concentration both directly and through the ensuing sodium load that increases renal sodium loss with a parallel, obligate renal water loss. 100-150ml boluses of 3% sodium chloride can be used to increase plasma sodium by some 2–4 mmol/L over the initial 2–4 hours reducing intra-cerebral pressure in the acute setting. Thereafter, the increase in plasma sodium should be limited to 10 mmol/L in the first 24 hours. Given the risk of over-correction, a pragmatic strategy is to aim for a rise of 6-8 mmol/L in first 24 hours.  After the first 24 hours, the rate of rise of sodium should be limited to no more than 8 mmol/L per day.

 

Avoidance of over-correction is critical. If over-correction does occur, it is important to seek expert advice and consider actively controlling the rate of rise of plasma sodium with hypotonic fluid. Hypertonic fluid should be stopped when the defined clinical target (such as cessation of seizures) or a sodium concentration of 130 mmol/L is reached, whichever is first. If hypertonic sodium chloride cannot be given safely, it should not be given.

 

Patients with Mild or Less Severe Symptoms and Signs

 

The clinician has time to make a diagnosis, identify and address contributing factors if possible, and introduce a cause-dependent intervention.  Importantly, the limits on rate of rise of sodium remain the same as for the patient with critical signs and symptoms. The majority of patients will have circulating volume depletion or SIAD. Where these are due to drug treatments, the removal of the causal agent may be sufficient to normalise sodium. There will be some cases where the causal agent cannot be removed or where there is an alternative diagnosis.  Plasma sodium may rise faster than expected due to ‘auto correction’ of hyponatremia when an underlying cause has simply been removed e.g. correction of glucocorticoid insufficiency or withdrawal of excess desmopressin. Osmotic demyelination can still occur in these circumstances and so care must be taken to control the rise of plasma sodium such that the rate remains within target.

 

Fluid Challenge in Hypovolemic Hyponatremia

 

Mild to moderate hypovolemia can be difficult to diagnose clinically and low urine sodium excretion (<20 or 30 mmol/L) may be unreliable as a diagnostic test in the face of diuretic use or renin-angiotensin system blockade. If volume depletion is suspected, a moderate intravenous fluid challenge with 0.5–1 L N-saline over 2–4 hours may be both diagnostic and therapeutic.

 

Fluid Restriction in SIAD

 

Fluid restriction of 0.5–1 L/day is a reasonable initial intervention when excess plasma water is suspected and when the clinical condition is not critical. In patients with primary polydipsia, reduction in fluid intake remains the most reasonable approach. All fluids need to be included in the restriction. As SIAD is associated with negative sodium balance, sodium intake needs to be maintained. Several days of restriction may be required before sodium levels rise and a negative fluid balance needs to be confirmed through appropriate monitoring.


Management of Persistent Hyponatremia

 

Hyponatremia may persist or recur after initial intervention. It is important that the differential diagnosis is reviewed and the basis for intervention reconsidered.

-  In SIAD, fluid restriction may be only partly effective or may prove non-sustainable

-  Chronic liver or cardiac dysfunction may persist

-  Drug therapy exacerbating hyponatremia may need to continue

 

Clinical decisions on further management have to balance the merits of incremental intervention with those of tolerating mild, persisting hyponatremia.  Treatment aimed at simply raising plasma sodium in the absence of signs or symptoms may be of limited benefit and it is important to consider the clinical ‘trade-offs’ that may be required.

 

Demeclocycline

 

Demeclocycline has been used in SIAD. It produces a form of nephrogenic diabetes insipidus and so increases renal water loss, even in the presence of high concentrations of AVP. Treatment is 600–1,200 mg/day in divided doses. There is a lag time of some 3–4 days in onset of action. Dose adjustment needs to take this into account. Photosensitive skin reactions and renal impairment are significant adverse effects and limit clinical utility.

 

Vasopressin Receptor Antagonists

 

Vasopressin receptor antagonists (Vaptans) are a rational approach to the management of SIAD. They are classified as either selective (V2-R specific) or non-selective (V2- and V1a antagonism). Both increase renal water excretion without a significant impact on renal electrolyte loss (aquaretic action).  To avoid precipitating a rapid rise in plasma Na+, fluid restriction should be relaxed if these drugs are introduced. They should not be used in profound hyponatremia or in patients with severe symptoms. Their role in the management of SIAD in the majority of patients remains unclear.

 

GUIDELINES

 

  1. Spasovski G, Vanholder R, Allolio B et al. Clinical practice guideline on diagnosis and treatment of hyponatremia. European Journal of Endocrinology 2014; 170: G1–G47
  2. Verbalis JG, Goldsmith SR, Greenberg A, Korzelius C, Schrier RW, Sterns RH, Thompson CJ. Diagnosis, evaluation and treatment of hyponatremia: expert panel recommendations. The American Journal of Medicine 2014;126: S1-S42  

 

REFERENCES

 

  1. Hannon MJ, Behan LA, Brien MMC, Tormey W, Ball S, Javadpour M, Sherlock M, Thompson CJ. Hyponatremia following mild/moderate subarachnoid haemorrhage is due to SIAD and glucocorticoid deficiency and not cerebral salt wasting. Journal of Clinical Endocrinology and Metabolism 2014; 99: 291-298
  2. Ward FL, Tobe SW, Naimark DMJ. The Role of Desmopressin in the Management of Severe, Hypovolemic Hyponatremia: A Single-Center, Comparative Analysis. Canadian Journal of Kidney Health and Disease 2018; 5: 1–8
  3. Ball SG. The Neurohypophysis: Endocrinology of Vasopressin and Oxytocin. In: De Groot LJ, Chrousos G, Dungan K, Feingold KR, Grossman A, Hershman JM, Koch C, Korbonits M, McLachlan R, New M, Purnell J, Rebar R, Singer F, Vinik A, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000-2017 Apr 22. PMID: 25905380

Benign Breast Disease in Women

ABSTRACT

Benign breast disease in women is a very common finding and results in a diagnosis in approximately one million women annually in the United States (1). An understanding of the hormonal and growth factor control of breast development and function is key to the rational and systematic evaluation and treatment of patients. A firm understanding of benign breast disease is important since sequential steps are necessary to distinguish lesions which impart a high risk of subsequent breast cancer from those which do not. This chapter will review the physiology of breast function, provide histologic examples of common lesions, and detail practical approaches to evaluation and treatment.  For complete coverage of all related areas of Endocrinology, please see our online FREE web-book, www.endotext.org.

 

BREAST PHYSIOLOGY IN WOMEN:

Hormones and growth factors act upon stromal and epithelial cells to regulate mammary gland development, maturation and differentiation (2). Broadly summarized, estrogen mediates development of ductal tissue; progesterone facilitates ductal branching and lobulo-alveolar development; and prolactin regulates milk protein production. At puberty, estradiol and progesterone levels increase to initiate breast development. A complex tree-like structure results and comprises 5 to 10 primary milk ducts originating at the nipple, 20 to 40 segmental ducts, and 10 to 100 sub-segmental ducts ending in glandular units called terminal duct lobular units (TDLUs) (3). During the menstrual cycle the increments in estrogen and progesterone stimulate cell proliferation during the luteal phase (Figure1). Cycle dependent apoptosis balances proliferation (4). In response to enhanced proliferation, the breast can increase by as much as 15% in size during the luteal phase.

Anatomic and histologic structures of the breast undergo substantial change during the period from early adolescence to menopause (5). The normal histologic appearance represents a spectrum ranging from a predominance of ducts, lobules, and intra- and inter-lobular stroma to patterns with a predominance of fibrous change and cyst formation, a process formerly called fibrocystic disease (Figure 2). The term “fibrocystic changes” is now preferred since up to 50 to 60 percent of normal women may have this pattern histologically (6). This new term implies that women with lumpy breasts or non-discrete nodules do not have breast disease. Importantly, fibrocystic changes detected clinically incur no increased risk of breast cancer.

 

Specific changes in the breast, relating to stromal, ductal and glandular tissue occur as a function of age. During the early reproductive years, stromal hyperplasia may occur and produces juvenile breast hypertrophy (7) or rarely, the more significant problems of unilateral or bilateral macromastia (enlargement of breast tissue beyond what is considered normal) (8). Changes in glandular and ductal tissue occur uncommonly. In the middle reproductive years, glandular breast tissue continues to undergo changes in response to cyclic increments in plasma levels of estradiol and progesterone and, if substantial, is called adenosis. Ductal changes remain uncommon while stromal hyperplasia may occur resulting in areas of ill-defined fullness (“lumpy-bumpy” consistency) on physical exam or in firm areas requiring biopsy.

 

 

Figure 1. Influence of the cycle phase on breast total labeling index (TLI) of women less than 34 years of age according to whether cycles were natural or regulated by oral contraceptives (OC). (Reprinted with permission from Going JJ, Anderson TJ, Battersby et al. Proliferative and secretory activity in human breast during natural and artificial menstrual cycles (Am. J Pathol. 130:193-204, 1988).Figure 1. Influence of the cycle phase on breast total labeling index (TLI) of women less than 34 years of age according to whether cycles were natural or regulated by oral contraceptives (OC). (Reprinted with permission from Going JJ, Anderson TJ, Battersby et al. Proliferative and secretory activity in human breast during natural and artificial menstrual cycles (Am. J Pathol. 130:193-204, 1988).

Figure 2 . Simplified anatomy of the female breast illustrating the major structural components of the breast, the anatomic location of various lesions, and the histology of those lesions and corresponding sites of origin of potential lesions.

Figure 2 . Simplified anatomy of the female breast illustrating the major structural components of the breast, the anatomic location of various lesions, and the histology of those lesions and corresponding sites of origin of potential lesions.

In the late reproductive period, glandular tissue may become hyperplastic with sclerosing adenosis or lobular hyperplasia. The hyperplastic glandular lesions may progress to palpable or mammographically detectable abnormalities requiring biopsy. Ductal tissue also may undergo hyperplastic change with an increase in number of ductal cells but without alterations in their appearance. This change, when associated with a 2% or greater prevalence of Ki67 positive cells, is associated with an approximately two fold increase of subsequent breast cancer (9). Further proliferative changes result in lobules approaching 100µ in diameter and called hyperplastic elongated lobular units (HELUs). With progression of the HELUs, atypical ductal or lobular hyperplasia, or ductal carcinoma in situ (DCIS) may ensue. The linear pathway illustrated in Figure 3 depicts this progression but, while considered by some to be overly simplistic, provides a useful framework. This orderly progression depends upon the number of acquired genetic mutations accumulated by clonal cells in the breast. This originally was suggested by loss of heterozygosity studies. Recent specific genetic data suggests that an average of 11 “driver” mutations and 100 bystander mutations are present in established invasive breast cancer (10). Based on the number of mutations required for cancer, current opinion suggests that breast cancer is a process that takes many years to develop with the age of menarche as the earliest factor influencing this process. The progression from the earliest neoplastic changes to invasive breast cancer is considered to take a median of 16 years with a range of 1 to 30 years based on doubling time. Careful assessment of serial mammograms allows an estimation of doubling times which range from 25 to more than 700 days (Figure 4).

Figure 3 Model of the linear progresion from normal, to hyperplastic enlogated lobular units ( HELU), to atypical ductal hyperplasia (ADH), to ductal carcinoma in situ ( DCIS), to occult invasive breast cancer ( IBC), to IBC that has exceeded detection threshold and can be diagnosed by mammogram. Kinetic modeling data suggest that it takes 10 to 20 years for the progression from atypical hyperplasia to a clinically detectable tumor

Figure 3 Model of the linear progresion from normal, to hyperplastic enlogated lobular units ( HELU), to atypical ductal hyperplasia (ADH), to ductal carcinoma in situ ( DCIS), to occult invasive breast cancer ( IBC), to IBC that has exceeded detection threshold and can be diagnosed by mammogram. Kinetic modeling data suggest that it takes 10 to 20 years for the progression from atypical hyperplasia to a clinically detectable tumor

Figure 4. Cumulative frequency distribution of the doubling times of occult breast cancer obtained by serial mammograms. Figure reproduced with the permision of the author and publisher.

Figure 4. Cumulative frequency distribution of the doubling times of occult breast cancer obtained by serial mammograms. Figure reproduced with the permision of the author and publisher.

After the onset of menopause, glandular tissue undergoes involution and stroma and fatty tissue (i.e. approximately 97%) replace glandular elements (12).  The degree of involution is inversely related to the risk of subsequent breast cancer. When comparing the lowest with the highest quartile of involution, the relative risk of subsequent breast cancer is more than 2 fold increased I (OR 2.44 , 95% confidence interval (0.96-6.19) (1).  With aging, the degree of involution increases (1), which contributes substantially to the decrease in breast density. On histologic examination, lesions appearing dense on mammography contain a larger than normal proportion of stromal and glandular tissue.

SPECIFIC BENIGN BREAST LESIONS:

A wide variety of benign breast lesions have been described and the histologic appearance fully characterized. On a practical basis, these can be subdivided into those associated with no substantial increased risk of breast cancer ( i.e. < 1.49%), those with an increase of 1.5-2% and those with a >2% increase (Table 1) (13) .

Table 1. Relative risk of breast cancer imparted by specific benign breast lesions (Data from Reference 13)

No Increased risk:

The more common lesions Included in this category are fibrocystic changes, periductal fibrosis, hamartomas, lipomas, phylloides tumors, and neurofibromas. Another lesion is duct ectasia (Figure 5), characterized by distention of subareolar ducts and presence within them of yellowish-orange material. Penetration of the duct wall by this material may produce acute inflammatory changes in the surrounding tissues. Histologically, crystalline oval and round structures thought to be lipid in origin are present. Resolution may occur but residual periductal fibrosis and nodule formation often persist. This lesion is thought to be more frequent in cigarette smokers and represents a more chronic and extensive inflammatory process. A less common lesion is diabetic mastopathy which consists of localized or diffuse areas of fibrosis occurring in patients with diabetes (14).

Figure 5. Histological appearance of duct ectasia showing the characteristic crystalline formation of the intraluminal contents.

Figure 5. Histological appearance of duct ectasia showing the characteristic crystalline formation of the intraluminal contents.

Abnormalities of ductal secretion may result in discharge of clear, cloudy, blue, green or black aqueous material. Stromal hyperplasia can result in nipple retraction or in palpable lesions requiring biopsy to distinguish from breast carcinoma. Histologically, the tissue may contain fibroblasts nearly exclusively or predominantly fibroblasts with admixture of glandular epithelium. Some degree of stromal hyperplasia may occur in 50-60% of normal women, particularly those in their middle and late reproductive periods. Traumatic lesions (hematomas and fat necrosis) and inflammatory conditions (granulomas and mastitis) are also not associated with an increased breast cancers (15).

 

1.5 To 2.0 Fold Increase In Relative Risk:

 

Fibroadenomas:
During the early reproductive period, glandular components of the breast may respond to cyclic hormonal stimuli in an exaggerated fashion with the development of single fibroadenomas. These consist of lobular units which grow to larger than normal size and contain both epithelial and stromal elements. Fibroadenomas range in size from slightly larger than a normal single lobular unit to larger, more discrete palpable lesions. When exceeding 5 cm, these are called giant fibroadenomas which are seen primarily at puberty or during pregnancy. The incidence of fibroadenomas peaks at age 20-24 (Figure 6). The prevalence on physical examination in young women is 2% in young women but 15-23% when evaluated prospectively at autopsy (16). When complex and containing cysts >3mm in diameter, sclerosing adenosis, epithelial calcification or papillary changes, these lesions are associated with an increased risk of breast cancer (17) (Table 1).

Figure 6. Incidence of fibrocystic breast changes and fibroadenoma by age group. (Reprinted with permission of the publisher and authors from Goehring C, Morabia A. Epidemiology of Bengin Breast Disease, with special attention to histologic types Epidemiologic Reviews 19:310-327, 1997) (16). Note that the term “fibrocystic disease” is used in the legend but this term has now been changed to “fibrocystic changes”.

Figure 6. Incidence of fibrocystic breast changes and fibroadenoma by age group. (Reprinted with permission of the publisher and authors from Goehring C, Morabia A. Epidemiology of Bengin Breast Disease, with special attention to histologic types Epidemiologic Reviews 19:310-327, 1997) (16). Note that the term “fibrocystic disease” is used in the legend but this term has now been changed to “fibrocystic changes”.

Other lesions:

Hyperplasia without atypia, papillomas, papillomatosis, radial scar, blunt duct adenosis, and sclerosing adenosis are also associated with an increased risk.

 

Greater Than A Two-Fold Risk:

 

Atypical hyperplasia

This lesion can be subdivided into atypical ductal hyperplasia (figure 7) and atypical lobular hyperplasia but both impart a similar increase in relative risk of breast cancer of 3 to 4 fold (18) (Table 1). In the first ten years after diagnosis of AH, the breast tumors occur predominantly in the same breast but subsequently, occur with equal frequency in the contralateral as in the ipsilateral breast (19). These observations suggest the AH is both a precursor lesion for breast cancer and a marker of an underlying predisposing condition termed a “field defect” or alternatively a “mutator phenotype (15) When the amount of hyperplastic tissue increases according to specific criteria (20) the lesions are no longer called benign but are classified as ductal carcinoma in situ. Recent emphasis has been directed toward identification of molecular genetic markers which could predict which patients with AH are at increased risk of developing breast cancer. Over-expression of HER-2/neu is one such marker (21). The presence of aberrant p53, p21, interleukin 6, and TNF alpha and aneuploidly on flow cytometery have also been reported as markers of increased risk (22).

Figure 7. Photomicrograph of atypical ductal hyperplasia histology reprinted from Hartmann LC et al (23).

Figure 7. Photomicrograph of atypical ductal hyperplasia histology reprinted from Hartmann LC et al (23).

Recent data from the Mayo Clinic report the absolute rates of breast cancer during prolonged 20+ year follow-up and the important role of multifocality (23;24). As shown in Figure 8, the risk approximates 24% with one lesion, 36% with two and 47% with 3 or more lesions. The increased risk with multifocality has been confirmed in an independent cohort follow at  Vanderbilt (18;19). Although previous studies indicated that family history added to this risk (18), the updated data suggest that neither family history or other risk factors independently contribute to breast cancer risk (23). It is important to emphasize the AH appears to impart a risk of breast cancer which is lower but may approach that from BrCa1 and 2 mutations. As noted above, the presence of atypical hyperplasia imparts a risk over time of both ipsilateral and contralateral breast cancer (Figure 9).

Figure 8. Incidence of breast cancer in cohort of patients with a histologic diagnosis of atypical hyperplasia. Top panel: mean (solid line) and 95% confidence limits (dashed lines). Bottom panel: Incidence in patients with 1, 2, or 3 or more independent lesions. Reprinted with the permission of the authors and publisher from Hartmann LC et al (23).

Figure 8. Incidence of breast cancer in cohort of patients with a histologic diagnosis of atypical hyperplasia. Top panel: mean (solid line) and 95% confidence limits (dashed lines). Bottom panel: Incidence in patients with 1, 2, or 3 or more independent lesions. Reprinted with the permission of the authors and publisher from Hartmann LC et al (23).

Figure 9. Ipsilateral versus contralateral distribution of breast cancers diagnosed over time in patients initially diagnosed with atypical hyperplasia. Reproduced with the permission of the authors and publishers from Hartmann LC et al (24).

Figure 9. Ipsilateral versus contralateral distribution of breast cancers diagnosed over time in patients initially diagnosed with atypical hyperplasia. Reproduced with the permission of the authors and publishers from Hartmann LC et al (24).

Lobular Carcinoma in situ:

Even though called lobular carcinoma in situ, this lesion is not generally considered to have reached the neoplastic stage but is analogous to atypical hyperplasia. This lesion imparts approximately a 10 fold   increase in relative risk of breast cancer developing over time. The subsequent breast cancer can occur in the same or contralateral breast; accordingly, this lesion is thought to represent a filed defect or mutator phenotype which increases breast cancer risk but the lesion itself is not a precursor of breast cancer.

 

 

Mammographic Density And Breast Cancer Risk:

The percent breast density on mammography correlates with breast cancer risk as shown on Figure 10. When comparing the lowest density category with highest, the relative risk is increased by 5.3 fold. Recent studies examined the histologic composition of dense and non-dense breast tissue. When dense lesions are biopsied and compared to areas of low density, they are found to contain a higher proportion of stroma and glandular tissue and lesser amount of fat (26). Notably, dense lesions contain more of the enzyme aromatase, when quantitated by an immunologic histologic score after staining with an aromatase monoclonal antibody (27). These findings are likely associated with a higher local production of estradiol and could explain the higher incidence of breast cancer with both of these lesions. Taking these observations together, determination of breast density assessed by Bi-RADS criteria or quantitated with a computer assisted method of determining breast density, provides the most powerful means of predicting risk of breast cancer over time (Figure 10).

Figure 10. The percentage of breast tissue which is dense on a mammogram is determined by computer assisted analysis and classified as none, <10%, 10-25%, 25-50%, 50-75%, >75%.The relative risk of breast cancer increases with each step of increased breast density. Adapted from that data of Boyd et al (25).

Figure 10. The percentage of breast tissue which is dense on a mammogram is determined by computer assisted analysis and classified as none, <10%, 10-25%, 25-50%, 50-75%, >75%.The relative risk of breast cancer increases with each step of increased breast density. Adapted from that data of Boyd et al (25).

ETIOLOGY OF BENIGN BREAST DISORDERS

 

Clinical observations in women receiving estrogens and anti-estrogens suggest that hormonal events play a role in the etiology of benign breast lesions. In post-menopausal women receiving estrogens ± progestins for >8 years, the prevalence of benign breast lesions increased by 1.7 fold (95 percent CI 1.06-2.72) (28). In the Women’s Health Initiative study (WHI), the use of estrogen plus progestin was associated with a 74% increase in the risk of benign proliferative breast disease [hazard ratio, 1.74; 95% confidence interval (CI), 1.35-2.25] (29). The anti-estrogen, tamoxifen, when used for breast cancer prevention, was associated with a 28 percent (RR 0.72, 95 percent Confidence Intervals 0.65-0.79) reduction in prevalence of benign breast lesions, including adenosis, cysts, duct ectasia, and hyperplasia (30).

 

Underlying and acquired genetic changes are also associated with benign breast lesions. Loss of heterozygosity (LOH), a finding caused by deletions of small segments of DNA (31;32) is commonly found in benign breast lesions. Women frequently have multi-focal lesions, each of which exhibit loss of heterozygosity (LOH) of differing regions of DNA. Women with BRCA1/2 mutations are found to have a high frequency of multiple benign or malignant breast lesions when bilateral mastectomy specimens are meticulously examined (33). These findings support the current theory of an underlying predisposition to mutations in some patients as the cause of multiple breast lesions (34). In the past, this phenomenon was termed a “field effect” and more recently, a “mutator phenotype” (34).

 

CLINICAL MANIFESTATIONS:

Clinical presentations of benign breast disease are divided into those with pain, lumps, or breast discharge (Table 2).

Breast Pain:

Cyclic breast pain

 

Usually occurs during the late luteal phase of the menstrual cycle, in association with the premenstrual syndrome or independently (35-40), and resolves at the onset of menses (35;36;38). A recent study in 1171 healthy American women indicated that 11% experience moderate to severe cyclic breast pain and 58%, mild discomfort (39). Breast pain interfered with usual sexual activity in 48% and with physical (37%), social (12%), and school (8%) activity in others. A role for caffeine, iodine deficiency, alterations in fatty acid levels in the breast, fat intake in the diet, and psychological factors in the etiology of breast pain remain unproven. Non-cyclic breast pain is unrelated to the menstrual cycle. Detection of focal tenderness is helpful diagnostically and suggests a tender cyst, rupture through the wall of an ectatic duct, or a particularly tender area of breast nodularity. Acute enlargement of cysts and periductal mastitis may cause severe, localized pain of sudden onset.

 

Non-breast pain:

When arising from the chest wall, pain may be mistakenly attributed to the breast. Pain localized to a limited area and characterized as burning or knife like in nature suggests this possibility. Several distinct subtypes can be distinguished including localized or diffuse lateral chest wall pain, radicular pain from cervical arthritis and Tietze’s syndrome or costochondritis. The method to distinguish between breast and non-breast pain by careful physical examination is illustrated on Figure 11, panels A-D.

Figure 11. A. Focal chest wall pain—lateral. The patient is turned 90º on her side so that breast tissue is no longer under the area of palpation. The index finger elicits a focal area of pain. B. Focal chest wall pain over costochondral junctions anteriorly. C. Diffuse lateral chest wall pain. With the patient turned over 90 degrees on her side, pain is elicited over a wider area of the chest wall. D. Verification that squeezing breast tissue does not elicit pain ensures that the pain is not related to the breast but represents chest wall pain.

Figure 11. A. Focal chest wall pain—lateral. The patient is turned 90º on her side so that breast tissue is no longer under the area of palpation. The index finger elicits a focal area of pain. B. Focal chest wall pain over costochondral junctions anteriorly. C. Diffuse lateral chest wall pain. With the patient turned over 90 degrees on her side, pain is elicited over a wider area of the chest wall. D. Verification that squeezing breast tissue does not elicit pain ensures that the pain is not related to the breast but represents chest wall pain.

Breast Nodule:

Proliferation of ductal or lobular tissue causes histologic changes that are manifested by the presence of palpable lumps or nodules. Patients often present with the finding of a new breast nodule on self-exam or are found to have a lump by their health care provider. Ninety percent of these new nodules in premenopausal women are benign and usually represent fibroadenomas in the early reproductive period (16). In the middle reproductive period, focal areas of fibrosis, hyperplasia, or cyst formation are more likely. In the later reproductive period, hyperplasia, cysts, and carcinoma in situ are more common.  Some lesions present with symptoms suggesting the cause. With duct ectasia, penetration of the duct wall by lipid material may be associated with a nodule exhibiting acute redness, and causing pain, and fever; after resolution, a subareolar nodule persists.

Other specific lesions present as lumps. These include multiple papillomas, sclerosing adenosis, and radial scars. Multiple papillomas may present as breast lumps, nodules on ultrasound, or may be the cause of bloody nipple discharge and can be seen on ductography. Sclerosing adenosis is a lobular lesion with increased fibrous tissue and interspersed glandular cells. It can present as a mass or a suspicious finding on mammograms. Radial scars are a pathologic diagnosis, usually diagnosed following mammography or palpation and then biopsy. Radial scars are characterized microscopically by a fibroelastic core with radiating ducts and lobules and impart a minimally increased risk of breast cancer similar to that of proliferative changes without atypia (41).

Nipple discharge:

6.8 percent of referrals to physicians for breast concerns result from this symptom. Although particularly distressing to the patient, only 5 percent are found to have serious underlying pathology. Age is an important factor with respect to risk of malignancy (42). In one series, 3 percent of women younger than age 40, 10 percent between 40 and 60 and 32 percent >60 with nipple discharge as their only symptom were found to have a malignancy.

 

A careful history characterizes breast discharge as either spontaneous or expressible. On examination, one can detect by careful inspection whether the discharge emanates from a single or multiple ducts. Nipple discharge can be divided into physiologic and pathologic types. Characteristics of physiologic discharge include non-spontaneous, multiple duct, bilateral, and non-bloody. Pathologic discharge is characterized as spontaneous, serous or bloody, usually unilateral and usually single duct. Reassuring characteristics are that it must be expressed; is green yellow, brown or milky; that it is bilateral and involves multiple ducts. Spontaneous discharge, whether serous or bloody, requires careful evaluation. A hemoccult card or urine dipstick can be used to test for occult blood if the discharge is spontaneous, unilateral, and from one duct. Cytologic examination is not recommended. Milky discharge (galactorrhea) should be evaluated with measurement of a serum prolactin level. If the discharge is physiologic and the patient is under 35, only reassurance is necessary. Screening mammogram is recommended for patients over 35 with physiologic discharge. Pathologic discharge requires diagnostic mammogram, galactography (Figure 12) (43), and referral to a surgeon.

Figure 12. Galactogram illustrating space occupying lesion. A catheter is inserted into the duct from which the bloody discharge emerges. Contrast material is then injected through the catheter. The various branches of the duct are outlined.

Figure 12. Galactogram illustrating space occupying lesion. A catheter is inserted into the duct from which the bloody discharge emerges. Contrast material is then injected through the catheter. The various branches of the duct are outlined.

The presence of crusting, scaling, and flaking of the nipple could be a manifestation of Paget’s disease of breast and underlying cancer or of dermatologic problems. The approach is to obtain a history of other dermatologic problems, or a history of change in soap or clothing. If absent, a diagnostic mammogram should be obtained if the patient is over 35. In a large recent series of 1251 patients with nipple discharge, 433 had unilateral discharge and 194 had no breast lump in association with this symptom. Of these, the lesions found included solitary papilloma (n=49), minimal breast cancer (n=20), fibrocystic disease(n=11), papillomatosis (n=7), lobular cancer (n=5) and duct ectasia (n=2)(42) (30A). For women with bloody discharge from a single duct, galactography is warranted. Filling defects can be due to intraductal papilloma, intraductal carcinoma, papillomatosis, debris, or air bubbles.

 

 

APPROACH TO THE PRACTICAL MANAGEMENT OF BENIGN BREAST DISEASE

 

A detailed history and physical exam systematically evaluates the entire breast and chest wall and focuses on areas involving the patient’s symptoms (Table 3). Diagnostic studies may then be ordered. For lumps, “The Triple Test” is recommended which includes palpation, imaging and percutaneous biopsy (either core or fine needle aspirate <FNA>). Mammography, often in conjunction with ultrasound examination (44-47) is required for evaluation of discrete palpable lesions in women over 35 whereas ultrasound provides an optional substitute in younger women (43). Round dense lesions on mammography often represent cysts which require only ultrasonography to distinguish them from solid lesions. Complex cysts containing both fluid and solid matter require biopsy.  For solid lesions, radiographically or ultrasonically directed core biopsy provides highly discriminative information regarding the presence or absence of malignancy. Core biopsy utilizes a large cutting needle deployed with a spring loaded, automated biopsy instrument and obtains tissue suitable for histologic analysis familiar to most pathologists. FNA frequently yields sufficient cellular material to allow adequate cytologic evaluation but requires an experienced cytopathologist. However, the amount of material obtained is insufficient to render a diagnosis in 35-47% of non-palpable lesions (48) and core biopsy is recommended. The exact role of MRI in evaluating breast lesions is currently being determined (49). Galactography (ductography) is useful for detection of focal lesions in a single duct. Cytology of nipple discharge is of limited value with the sensitivity of detecting malignancy only 35 to 47 percent (50).

Diagnostic Procedures

Ideally a team including a radiologist experienced in mammography, ultrasound, MRI and core needle biopsy as well as an internist, gynecologist, or surgeon with expertise in breast diseases should be involved in the evaluation of patients with breast disorders. MRI mammography, ductography, or ultrasound may be utilized (Figures 12 and 13). Information to be obtained by a focused history and physical examination are outlined on Table 3. The method of documenting whether breast pain is chest wall related is illustrated on Figure 11 A-D. Imaging has become an integral part of the management of benign breast disorders.

 

Imaging studies:

Mammography is useful for evaluation of palpable lesions, particularly in those over 35. Digital mammography is preferred because of its ability to penetrate through dense breast tissue which is commonly found in younger women. Ultrasound is often used as initial evaluation of a palpable mass in women under age 35. If a simple cyst is present, no further evaluation is necessary (Figure 13). If not, mammography may also be necessary to fully evaluate the lump. If the mass has findings suggestive of a fibroadenoma by ultrasound and mammography, short term follow-up and re-imaging can be considered (usually performed in 6 months). Experts are divided as to the necessity to biopsy all fibroadenomas. MRI is more sensitive than digital mammography but false positives are more common (49). Routine yearly MRI is now recommended for women whose lifetime risk of breast cancer is > 20%. As an illustration of its sensitivity, 3% of the contralateral breasts in women with diagnosed breast cancer are found to have a second lesion in the opposite breast when examined by MRI (49).

Figure 13 Upper panel illustrates by ultrasound a non-dense black area representing cyst fluid. The lower panel is the corresponding area on mammogram showing a dense area. With the combination of mammogram and ultrasound, the lesion can be shown to be a cyst.

Figure 13 Upper panel illustrates by ultrasound a non-dense black area representing cyst fluid. The lower panel is the corresponding area on mammogram showing a dense area. With the combination of mammogram and ultrasound, the lesion can be shown to be a cyst.

Findings On Imaging Studies

 

Fibrocystic change typically presents on mammogram as round or oval, well defined masses that can be subsequently shown to represent cysts on ultrasound (Figure 13). Diffusely scattered dystrophic calcifications may also be found on the mammogram. Consequently, the goal of mammographic evaluation is to provide reassurance to the patient and physician that the risk of neoplasm is low. Aspiration of cysts is usually necessary only in a those cases where the mass does not fulfill all criteria for a simple cyst or if the cyst is painful. Biopsy may be necessary to confirm the benign nature of calcifications, particularly if clustered, linear or variously shaped.

For round masses or round calcifications on a first mammogram, the risk of cancer is less than 2%, and repeat mammography in 6 months is recommended. These lesions are termed “probably benign” using the lexicon terminology required by the Mammography Quality Standards Act (in the USA). If the risk is believed to be greater, core biopsy is recommended. Stereotactically directed core biopsy is ideal for evaluation of calcifications and provides highly discriminative information regarding the presence or absence of malignancy. If this technique is not available, insertion of a wire into the lesion radiographically followed by surgical excision or mere removal of a palpable lesion is warranted.

 

CLINICAL GUIDELINES FOR EVALUATION OF NODULES AND DISCHARGE

 

Careful examination distinguishes solitary, discrete, dominant, persistent masses from vague nodularity and thickening. Practice Guidelines of the Society of Surgical Oncology (51) recommend the following evaluation: In women less than age 35, all dominant discrete palpable lesions require referral to a surgeon. If vague nodularity, thickening or asymmetrical nodularity is present, the examination is repeated at midcycle after one or two menstrual cycles. If the abnormality resolves, the patient is reassured and if not, referred to a surgeon. Breast imaging may be appropriate. In women > age 35 with a dominant mass, a diagnostic mammogram (and frequently a sonogram) (44-47) is obtained and the patient referred to a surgeon. With vague nodularity or thickening, one obtains a mammogram with repeat physical exam at mid-cyle 1 to 2 months later and refers to surgeon if the abnormality persists. Post-menopausal women are referred for surgical consultation after a mammogram. For gross cysts (i.e. >4 cm), the guidelines suggest needle aspiration with repeat imaging within six months. If the aspirated fluid does not contain blood, the fluid is discarded without further histologic analysis unless the cyst contains solid components (i.e. complex cyst). If the fluid contains blood or if the cyst is complex, the, fluid is sent for cytology and consultation from a surgeon requested. With persistent refilling of the same cyst after aspiration, surgical consultation is warranted.

 

Usual practice requires “the triple test” with palpation, mammography (often in conjunction with ultrasonography) and biopsy in women over age 35 with dominant masses. When mammography is negative but a dominant mass is present, biopsy is required to rule out malignancy since lobular carcinoma may not be seen on mammograms. In those younger, mammography may be omitted if ultrasound and biopsy yield definitive information. Many experts omit biopsy in younger women with lesions characteristic of fibroadenoma on ultrasound and elect to follow carefully with serial ultrasounds at six monthly intervals for two years and yearly thereafter. Since careful studies have shown that a lesion appearing benign on mammography and ultrasound is benign >99 percent of the time, clinical judgment may allow follow-up without biopsy in experienced hands (44-47). However, other experienced surgeons disagree and believe that all fibroadenomas require diagnostic core biopsy or FNA and especially in BRCA mutation carriers in whom medullary cancer may be found. Biopsy confirmation of a fibroadenoma eliminates the need for serial ultrasounds. For those with a diagnosis of ADH (Atypical Ductal Hyperplasia) on FNA or core biopsy, excisional biopsy is then required since more complete resection often changes the diagnosis to DCIS.

 

Breast discharge is evaluated according to the algorithm illustrated below on Figure 14. Careful attention to several factors are necessary including determination whether the discharge arises from one duct or multiple ducts, is bloody, or is milky.

Figure 14. Algorithm to assess breast discharge.

Figure 14. Algorithm to assess breast discharge.

TREATMENT OF BENIGN BREAST DISEASE IN WOMEN

 

The initial step in evaluating pain is to distinguish true breast pain from chest wall pain (Figure 11). Several well designed, randomized, controlled, double blind, cross over trials have validated the efficacy of medical therapy for cyclic mastalgia. Based upon these studies, we categorize therapies as definitely effective, definitely ineffective, possibly effective, and insufficiently studied. For classification as definitely effective, two or more randomized trials are required. For the category, possibly effective, one randomized trial must be positive in some respect but others may be negative. For the category definitely ineffective, prospective trials must be uniformly negative. For the category, insufficiently studied, only one randomized trial, either negative or positive is available. For full details and references see Table 162-1 in Endocrinology, Fourth Edition, LJ DeGroot and JL Jameson, editors, WB Saunders Company, Philadelphia publishers; Chapter 162 Benign Breast Disorders, RJ Santen page 2194)

 

Danazol, bromocriptine, and tamoxifen have been proven to be effective (52-55) (Figure 15). Linoleic acid in the form of evening primrose oil has been shown effective in two randomized trials but not in the third, the largest trial. Its role in treatment therefore remains uncertain (56-58) . Vitamin E is considered definitely ineffective and iodine and vaginal progesterone possibly effective. Medroxyprogesterone acetate, caffeine avoidance, and progesterone have not been sufficiently studied. Several other therapies have not been examined in randomized controlled trials but are likely to be beneficial since they are based upon physiologic principles. For example, precise fitting of a bra to provide support for pendulous breasts has been reported to relieve pain in observational studies. GnRH agonist analogues are used to lower LH, FSH, and estradiol levels and to create a temporary post-menopausal state (59;60). Onset of menopause is known to reduce the frequency of breast pain. This therapy is reserved for patients in whom all other measures fail and the pain is considered severe. Reduction of the dosage of estrogens in post-menopausal women or addition of an androgen to estrogen replacement therapy (e.g. Estrotestâ tablets) appears to be beneficial in reducing breast pain (personal observations of author).

Figure 15. Relative efficacy of agents to treat breast pain. These data are from the Breast Clinic in Cardiff, Wales and represent observational studies and not randomized, controlled efficacy trails.

Figure 15. Relative efficacy of agents to treat breast pain. These data are from the Breast Clinic in Cardiff, Wales and represent observational studies and not randomized, controlled efficacy trails.

Relative efficacy of effective therapies:

No large randomized, controlled studies have compared the relative efficacy of danazol, bromocriptine, evening primrose oil and tamoxifen. Figure 15 rank orders them according to efficacy based upon data from individual reports from the same clinic. Minimal data are available from clinical trials which involve direct head to head comparisons. It should be noted that overall responses to danazol, bromocriptine and evening primrose oil are lower in those with non-cyclic pain than those with cyclic pain. However, not all studies have carefully excluded patients with non-breast pain and therefore conclusions regarding non-cyclic pain should be considered tentative. The approach to non-breast pain is outlined in Figure 16.

Figure 16. Algorithm to treat non-breast pain

Figure 16. Algorithm to treat non-breast pain

WOMEN AT HIGH RISK FOR BREAST CANCER

 

A major consideration for women who present with breast problems is whether they have a higher than normal risk of developing breast cancer. Certain breast lesions such as fibrocystic changes are associated with no increased risk of subsequent breast cancer (Table I). A 1.5 to 2-fold greater risk of development of breast cancer over a 20 year period of follow-up occurs with proliferative lesions including ductal hyperplasia, lobular hyperplasia without atypia, sclerosing adenosis, diffuse papillomatosis and complex fibroadenomas. A recent report also suggested that radial scars increase relative risk by 1.8, a risk similar to that found in proliferative disease without atypia. It should be noted that the Gail model for assessing breast cancer risk, which is based predominantly on reproductive factors, underestimates the long term risk of breast cancer in women with benign breast disease. On the other hand, the five year prediction is more accurate (61).

 

The presence of dense breast tissue on mammography has also been reported to be a predictor of increased incidence of breast cancer (Figure 10).Two components of this finding must be considered: one, the presence of high breast density makes it more difficult to read mammograms and masks the sensitivity of finding a breast cancer initially but identifies it later and two, there is an increased risk of breast cancer associated with increased breast density. With long tem follow-up studies, masking is not the explanation for the increased breast cancer risk (62).

 

According to classic twin studies, heritability accounts for approximately 60% of the variation in breast density (63;64). Breast cancer risk is also increased in association with high plasma estradiol and testosterone levels in postmenopausal women (62;65) and 20 kg or more weight gain (66) in the pre-menopausal years.

 

Another risk factor is use of hormone replacement therapy. Current data from the Women’s Health Initiative suggests a 26% increase in relative risk of breast cancer with conjugated equine estrogen (CEE) when combined with medroxyprogesterone acetate when used for an average of 7.1 years. (67). This risk is probably increased further in women starting this therapy shortly after the menopause (i.e. a short gap between onset of menopause and start of menopausal hormone therapy)(68). Starting this therapy a long time after experiencing menopause (long gap) is associated with a lesser relative risk (68). The use of estrogen alone in the WHI was associated with a trend toward reduction of risk of breast cancer at five years and a statistically significant reduction in those adhering to therapy (68). In a follow up report, after 5 years of use and 8 additional follow-up years, the reported decrease of 23% was statistically significant (69) (manson). Available data suggest that the effects of menopausal hormone therapy in the WHI is a class effect and not related to the specific type of estrogen or progestin. One study, however, suggests that use of crystalline progesterone as the progestogen is associated with a lesser risk than use of medroxyprogesterone acetate (70).

 

To aid in assessing breast cancer risk, a questionnaire developed by Gail, utilizes answers to 7 questions to calculate the 5 year and lifetime risk of developing breast cancer (71). This model has recognized deficiencies in that it does not consider second degree relatives with breast cancer, proliferative lesions of breast other than ADH, alcohol intake, obesity, or birth control pill and menopausal hormone therapy (MHT) use. Nonetheless, the Gail model has been prospectively validated in over 6000 women followed for an average of 4.5 years. A more recently developed model, the Tyrer–Cuzick (IBIS II) model (72) incorporates second degree relatives, obesity and use of MHT into its risk calculations and provides more robust information than the Gail model.

 

BREAST CANCER PREVENTION

 

Patients with benign breast lesions imparting an increased risk of breast cancer can be offered tamoxifen (or raloxifene) as a prevention strategy. The risk of breast cancer is determined using the Gail or Tyrer-Cuzick model and the benefits versus risks of tamoxifen evaluated. Risk factors not included in the Gail or Claus models include degree of breast density, plasma free estradiol levels, bone density, weight gain after menopause, and waist-hip ratio (25;65;66;73). Current recommendations suggest that women with a five year risk of breast cancer of over 1.67 percent and no contraindications to tamoxifen should be informed about the possibility of taking tamoxifen for five years (74-77). A recent overview has shown a 38 percent reduction of the relative risk of breast cancer with tamoxifen but benefits may be offset by increased risks of thromboembolic phenomena, endometrial cancer, and maturation of cataracts (78). The Star trial addressed whether raloxifene might be preferable to tamoxifen and (79) demonstrated relative equivalence. However, of interest was the fact that tamoxifen prevented more non-invasive breast cancers than did raloxifene (79). More intensive and frequent screening with multi-modality imaging (i.e. digital or standard mammography plus ultrasound or MRI) may be required in high risk patients. Atypical hyperplasia lesions appear to be more amenable to breast cancer prevention as reviewed from non-head-to-head studies showing prevention ranging from 62 to 75% reduction in four separate randomized studies (23) when compared to a 38% reduction in women selected to be at high risk based on reproductive factors (78).

 

REFERENCE LIST

 

(1)   Figueroa JD, Pfeiffer RM, Brinton LA, Palakal MM, Degnim AC, Radisky D et al. Standardized measures of lobular involution and subsequent breast cancer risk among women with benign breast disease: a nested case-control study. Breast Cancer Research & Treatment 2016;159(1):163-172.

(2)   Russo J. Hormonal control of breast development. In: DeGroot LJ, Jameson JL, editors. Endocrinology. fourth edition ed. Philadelphia, PA: WB Saunders; 2001. p. 2181-2188.

(3)   Osborne MP. Breast anatomy and development. In: Harris JR, Osborne CK, Morrow M, Lippman ME, editors. Diseases of the Breast. Philadelphia: Lippincott Williams and Wilkins; 2000. p. 1-13.

(4)   Going JJ, Anderson TJ, Battersby S, MacIntyre CC. Proliferative and secretory activity in human breast during natural and artificial menstrual cycles. American Journal of Pathology 1988;130(1):193-204.

(5)   Hughes LE, Mansel RE, Webster DJ. Aberrations of normal development and involution (ANDI): a new perspective on pathogenesis and nomenclature of benign breast disorders. Lancet 1987;2:1316-1319.

(6)   Love SM, Gelman RS, Silen W. Sounding board. Fibrocystic "disease" of the breast--a nondisease? New England Journal of Medicine 1982;307(16):1010-1014.

(7)   Pearlman MD, Griffin JL. Benign breast disease. [Review] [30 refs]. Obstetrics & Gynecology 2010;116(3):747-758.

(8)   Baker SB, Burkey BA, Thornton P, LaRossa D. Juvenile gigantomastia: presentation of four cases and review of the literature. [Review] [16 refs]. Annals of Plastic Surgery 525;46(5):517-525.

(9)   Huh SJ, Oh H, Peterson MA, Almendro V, Hu R, Bowden M et al. The Proliferative Activity of Mammary Epithelial Cells in Normal Tissue Predicts Breast Cancer Risk in Premenopausal Women. Cancer Research 2016;76(7):1926-1934.

(10)   Wood LD, Parsons DW, Jones S, Lin J, Sjoblom T, Leary RJ et al. The genomic landscapes of human breast and colorectal cancers.[see comment]. Science 2007;318(5853):1108-1113.

(11)   Bailey SL, Sigal BM, Plevritis SK. A simulation model investigating the impact of tumor volume doubling time and mammographic tumor detectability on screening outcomes in women aged 40-49 years. Journal of the National Cancer Institute 2010;102(16):1263-1271.

(12)   Radisky DC, Visscher DW, Frank RD, Vierkant RA, Winham S, Stallings-Mann M et al. Natural history of age-related lobular involution and impact on breast cancer risk. Breast Cancer Research & Treatment 2016;155(3):423-430.

(13)   Dyrstad SW, Yan Y, Fowler AM, Colditz GA. Breast cancer risk associated with benign breast disease: systematic review and meta-analysis. [Review]. Breast Cancer Research & Treatment 2015;149(3):569-575.

(14)   Kudva YC, Reynolds C, O'Brien T, Powell C, Oberg AL, Crotty TB. "Diabetic mastopathy," or sclerosing lymphocytic lobulitis, is strongly associated with type 1 diabetes. Diabetes Care 2002;25(1):121-126.

(15)   Santen RJ, Mansel R. Benign breast disorders. [Review] [89 refs]. New England Journal of Medicine 2005;353(3):275-285.

(16)   Goehring C, Morabia A. Epidemiology of benign breast disease, with special attention to histologic types. Epidemiologic Reviews 1997;19(2):310-327.

(17)   Nassar A, Visscher DW, Degnim AC, Frank RD, Vierkant RA, Frost M et al. Complex fibroadenoma and breast cancer risk: a Mayo Clinic Benign Breast Disease Cohort Study. Breast Cancer Research & Treatment 2015;153(2):397-405.

(18)   Hartmann LC, Sellers TA, Frost MH, Lingle WL, Degnim AC, Ghosh K et al. Benign breast disease and the risk of breast cancer.[see comment]. New England Journal of Medicine 2005;353(3):229-237.

(19)   Degnim AC, Visscher DW, Berman HK, Frost MH, Sellers TA, Vierkant RA et al. Stratification of breast cancer risk in women with atypia: a Mayo cohort study.[see comment]. Journal of Clinical Oncology 2007;25(19):2671-2677.

(20)   Page DL, Dupont WD, Rogers LW, Rados MS. Atypical hyperplastic lesions of the female breast. A long-term follow-up study. Cancer 1985;55(11):2698-2708.

(21)   Stark A, Hulka BS, Joens S, Novotny D, Thor AD, Wold LE et al. HER-2/neu amplification in benign breast disease and the risk of subsequent breast cancer. Journal of Clinical Oncology 2000;18(2):267-274.

(22)   Garcia-Tunon I, Ricote M, Ruiz A, Fraile B, Paniagua R, Royuela M. Role of tumor necrosis factor-alpha and its receptors in human benign breast lesions and tumors (in situ and infiltrative). Cancer Science 2006;97(10):1044-1049.

(23)   Hartmann LC, Degnim AC, Santen RJ, Dupont WD, Ghosh K. Atypical hyperplasia of the breast--risk assessment and management options. New England Journal of Medicine 2015;372(1):78-89.

(24)   Hartmann LC, Radisky DC, Frost MH, Santen RJ, Vierkant RA, Benetti LL et al. Understanding the premalignant potential of atypical hyperplasia through its natural history: a longitudinal cohort study. Cancer Prevention Research 2014;7(2):211-217.

(25)   Boyd NF, Lockwood GA, Byng JW, Tritchler DL, Yaffe MJ. Mammographic densities and breast cancer risk. Cancer Epidemiology, Biomarkers & Prevention 1998;7(12):1133-1144.

(26)   Ghosh K, Brandt KR, Reynolds C, Scott CG, Pankratz VS, Riehle DL et al. Tissue composition of mammographically dense and non-dense breast tissue. Breast Cancer Research & Treatment 2012;131(1):267-275.

(27)   Vachon CM, Sasano H, Ghosh K, Brandt KR, Watson DA, Reynolds C et al. Aromatase immunoreactivity is increased in mammographically dense regions of the breast. Breast Cancer Research & Treatment 2011;125(1):243-252.

(28)   Rohan TE, Miller AB. Hormone replacement therapy and risk of benign proliferative epithelial disorders of the breast. European Journal of Cancer Prevention 1999;8(2):123-130.

(29)   Rohan TE, Negassa A, Chlebowski RT, Lasser NL, McTiernan A, Schenken RS et al. Estrogen plus progestin and risk of benign proliferative breast disease. Cancer Epidemiology, Biomarkers & Prevention 2008;17(9):2337-2343.

(30)   Tan-Chiu E, Wang J, Costantino JP, Paik S, Butch C, Wickerham DL et al. Effects of tamoxifen on benign breast disease in women at high risk for breast cancer. Journal of the National Cancer Institute 2003;95(4):302-307.

(31)   Allred DC, Mohsin SK. Biological features of human pre-malignant breast disease. In: Harris JR, Osborne CK, Morrow M, Lippman ME, editors. Diseases of the Breast. 2nd Edition ed. Philadelphia: Lippincott Williams and Wilkins; 2000. p. 355-366.

(32)   O'Connell P, Pekkel V, Fuqua SA, Osborne CK, Clark GM, Allred DC. Analysis of loss of heterozygosity in 399 premalignant breast lesions at 15 genetic loci. Journal of the National Cancer Institute 1998;90(9):697-703.

(33)   Hoogerbrugge N, Bult P, Widt-Levert LM, Beex LV, Kiemeney LA, Ligtenberg MJ et al. High prevalence of premalignant lesions in prophylactically removed breasts from women at hereditary risk for breast cancer. Journal of Clinical Oncology 2003;21(1):41-45.

(34)   Lundin C, Mertens F. Cytogenetics of benign breast lesions. Breast Cancer Research & Treatment 1998;51(1):1-15.

(35)   Khan SA, Apkarian AV. The characteristics of cyclical and non-cyclical mastalgia: a prospective study using a modified McGill Pain Questionnaire. Breast Cancer Research & Treatment 2002;75(2):147-157.

(36)   Kessel B. Premenstrual syndrome. Advances in diagnosis and treatment. Obstetrics & Gynecology Clinics of North America 2000;27(3):625-639.

(37)   Goodwin PJ, Neelam M, Boyd NF. Cyclical mastopathy: a critical review of therapy. British Journal of Surgery 1988;75(9):837-844.

(38)   Goodwin PJ, Miller A, Del Giudice ME, Ritchie K. Breast health and associated premenstrual symptoms in women with severe cyclic mastopathy. American Journal of Obstetrics & Gynecology 1997;176(5):998-1005.

(39)   Ader DN, Shriver CD. Cyclical mastalgia: prevalence and impact in an outpatient breast clinic sample. Journal of the American College of Surgeons 1997;185(5):466-470.

(40)   Ader DN, South-Paul J, Adera T, Deuster PA. Cyclical mastalgia: prevalence and associated health and behavioral factors. Journal of Psychosomatic Obstetrics & Gynecology 2001;22(2):71-76.

(41)   Berg JC, Visscher DW, Vierkant RA, Pankratz VS, Maloney SD, Lewis JT et al. Breast cancer risk in women with radial scars in benign breast biopsies. Breast Cancer Research & Treatment 2008;108(2):167-174.

(42)   Seltzer MH, Perloff LJ, Kelley RI, Fitts WT, Jr. The significance of age in patients with nipple discharge. Surgery, Gynecology & Obstetrics 1970;131(3):519-522.

(43)   Smith GE, Burrows P. Ultrasound diagnosis of fibroadenoma - is biopsy always necessary?[see comment]. Clinical Radiology 516;63(5):511-515.

(44)   Shetty MK, Shah YP, Sharman RS. Prospective evaluation of the value of combined mammographic and sonographic assessment in patients with palpable abnormalities of the breast. Journal of Ultrasound in Medicine 2003;22(3):263-268.

(45)   Moy L, Slanetz PJ, Moore R, Satija S, Yeh ED, McCarthy KA et al. Specificity of mammography and US in the evaluation of a palpable abnormality: retrospective review. Radiology 2002;225(1):176-181.

(46)   Soo MS, Rosen EL, Baker JA, Vo TT, Boyd BA. Negative predictive value of sonography with mammography in patients with palpable breast lesions. AJR American Journal of Roentgenology 2001;177(5):1167-1170.

(47)   Flobbe K, Bosch AM, Kessels AG, Beets GL, Nelemans PJ, von Meyenfeldt MF et al. The additional diagnostic value of ultrasonography in the diagnosis of breast cancer. Archives of Internal Medicine 2003;163(10):1194-1199.

(48)   Venta LA. Image guided biopsy of non-palpable breast lesions. Harris JR LM, Morrow M OCe, editors. Diseases of the Breast second edition. 149-164. 2000. Philadelphia, Lippincott Williams and Wilkins.

Ref Type: Generic

(49)   Smith RA. The evolving role of MRI in the detection and evaluation of breast cancer.[comment]. New England Journal of Medicine 2007;356(13):1362-1364.

(50)   Ambrogetti D, Berni D, Catarzi S, Ciatto S. The role of ductal galactography in the differential diagnosis of breast carcinoma. Radiologia Medica 1996;91(3):198-201.

(51)   Morrow M, Bland KI, Foster R. Breast cancer surgical practice guidelines. Society of Surgical Oncology practice guidelines. Oncology (Huntington) 1997;11(6):877-881.

(52)   Fentiman IS, Caleffi M, Brame K, Chaudary MA, Hayward JL. Double-blind controlled trial of tamoxifen therapy for mastalgia. Lancet 1986;1(8476):287-288.

(53)   Messinis IE, Lolis D. Treatment of premenstrual mastalgia with tamoxifen. Acta Obstetricia et Gynecologica Scandinavica 1988;67(4):307-309.

(54)   O'Brien PM, Abukhalil IE. Randomized controlled trial of the management of premenstrual syndrome and premenstrual mastalgia using luteal phase-only danazol. American Journal of Obstetrics & Gynecology 1999;180(1):1-23.

(55)   Millet AV, Dirbas FM. Clinical management of breast pain: a review. Obstetrical & Gynecological Survey 2002;57(7):451-461.

(56)   Pashby NL, Mansel RE, Hughes LE, Hanslip JI, Preece P. A clinical trial of evening primrose oil in mastalgi. British Journal of Surgery 1981;68:801.

(57)   Preece PE, Hanslip JI, Gilbert L. Evening primrose oil ( Efamol) for mastalgia. In: Horrobin DF, editor. Clinical uses of essential fatty acid. Montreal: Eden; 1982. p. 147-154.

(58)   Blommers J, de Lange-De Klerk ES, Kuik DJ, Bezemer PD, Meijer S. Evening primrose oil and fish oil for severe chronic astalgia: a randomized, double-blind, controlled trial. American Journal of Obstetrics & Gynecology 2002;187(5):1389-1394.

(59)   Hamed H, Caleffi M, Chaudary MA, Fentiman IS. LHRH analogue for treatment of recurrent and refractory mastalgia. Annals of the Royal College of Surgeons of England 1990;72(4):221-224.

(60)   Mansel RE, Goyal A, Preece P, Leinster S, Maddox PR, Gateley C et al. European randomized, multicenter study of goserelin (Zoladex) in the management of mastalgia. American Journal of Obstetrics & Gynecology 2004;191(6):1942-1949.

(61)   Tarabishy Y, Hartmann LC, Frost MH, Maloney SD, Vierkant RA, Pankratz VS. Performance of the Gail model in individual women with benign breast disease. Journal of Clinical Oncology 2011;29(15_suppl):1525.

(62)   Santen RJ, Boyd NF, Chlebowski RT, Cummings S, Cuzick J, Dowsett M et al. Critical assessment of new risk factors for breast cancer: considerations for development of an improved risk prediction model. [Review] [95 refs]. Endocrine-Related Cancer 2007;14(2):169-187.

(63)   Boyd NF, Jensen HM, Cooke G, Han HL, Lockwood GA, Miller AB. Mammographic densities and the prevalence and incidence of histological types of benign breast disease. Reference Pathologists of the Canadian National Breast Screening Study. European Journal of Cancer Prevention 2000;9(1):15-24.

(64)   Byrne C, Schairer C, Brinton LA, Wolfe J, Parekh N, Salane M et al. Effects of mammographic density and benign breast disease on breast cancer risk (United States). Cancer Causes & Control 2001;12(2):103-110.

(65)   Key T, Appleby P, Barnes I, Reeves G, Endogenous Hormones and Breast Cancer Collaborative Group. Endogenous sex hormones and breast cancer in postmenopausal women: reanalysis of nine prospective studies. Journal of the National Cancer Institute 2002;94(8):606-616.

(66)   Hulka BS, Moorman PG. Breast cancer: hormones and other risk factors. Maturitas 1928;38(1):103-113.

(67)   Chlebowski RT, Hendrix SL, Langer RD, Stefanick ML, Gass M, Lane D et al. Influence of estrogen plus progestin on breast cancer and mammography in healthy postmenopausal women: the Women's Health Initiative Randomized Trial.[see comment]. JAMA 2003;289(24):3243-3253.

(68)   Prentice RL, Manson JE, Langer RD, Anderson GL, Pettinger M, Jackson RD et al. Benefits and risks of postmenopausal hormone therapy when it is initiated soon after menopause.[see comment]. American Journal of Epidemiology 2009;170(1):12-23.

(69)   Manson JE, Chlebowski RT, Stefanick ML, Aragaki AK, Rossouw JE, Prentice RL et al. Menopausal hormone therapy and health outcomes during the intervention and extended poststopping phases of the Women's Health Initiative randomized trials. JAMA 2013;310(13):1353-1368.

(70)   Fournier A, Mesrine S, Boutron-Ruault MC, Clavel-Chapelon F. Estrogen-progestagen menopausal hormone therapy and breast cancer: does delay from menopause onset to treatment initiation influence risks?[see comment]. Journal of Clinical Oncology 2009;27(31):5138-5143.

(71)   Gail MH, Brinton LA, Byar DP, Corle DK, Green SB, Schairer C et al. Projecting individualized probabilities of developing breast cancer for white females who are being examined annually. Journal of the National Cancer Institute 1989;81(24):1879-1886.

(72)   Tyrer J, Duffy SW, Cuzick J. A breast cancer prediction model incorporating familial and personal risk factors.[see comment][erratum appears in Stat Med. 2005 Jan 15;24(1):156]. Statistics in Medicine 2004;23(7):1111-1130.

(73)   Kuller LH, Cauley JA, Lucas L, Cummings S, Browner WS. Sex steroid hormones, bone mineral density, and risk of breast cancer. Environmental Health Perspectives 1997;105:Suppl-9.

(74)   Chlebowski RT, Collyar DE, Somerfield MR, Pfister DG. American Society of Clinical Oncology technology assessment on breast cancer risk reduction strategies: tamoxifen and raloxifene. Journal of Clinical Oncology 1999;17(6):1939-1955.

(75)   Gail MH, Costantino JP, Bryant J, Croyle R, Freedman L, Helzlsouer K et al. Weighing the risks and benefits of tamoxifen treatment for preventing breast cancer. Journal of the National Cancer Institute 1999;91(21):1829-1846.

(76)   Levine M, Moutquin JM, Walton R, Feightner J, Canadian Task Force on Preventive Health Care and the Canadian Breast Cancer Initiative's Steering Committee on Clinical Practice Guidelines for the Care and Treatment of Breast Cancer. Chemoprevention of breast cancer. A joint guideline from the Canadian Task Force on Preventive Health Care and the Canadian Breast Cancer Initiative's Steering Committee on Clinical Practice Guidelines for the Care and Treatment of Breast Cancer. CMAJ Canadian Medical Association Journal 2001;164(12):1681-1690.

(77)   Chlebowski RT, Col N, Winer EP, Collyar DE, Cummings SR, Vogel VG, III et al. American Society of Clinical Oncology technology assessment of pharmacologic interventions for breast cancer risk reduction including tamoxifen, raloxifene, and aromatase inhibition. Journal of Clinical Oncology 2002;20(15):3328-3343.

(78)   Cuzick J, Powles T, Veronesi U, Forbes J, Edwards R, Ashley S et al. Overview of the main outcomes in breast-cancer prevention trials. Lancet 2003;361(9354):296-300.

(79)   Vogel VG. The NSABP Study of Tamoxifen and Raloxifene (STAR) trial. [Review] [35 refs]. Expert Review of Anticancer Therapy 2009;9(1):51-60.