throbber
Treatment of Cusn/ng’s disease
`
`223:2
`
`R1 9—R39
`
`This material may be protected by Copyright law [Title 17 US. Code]
`
`Treatment of Cushing’s disease:
`
`a mechanistic update
`
`Danlel Cuevas-Ramos"2 and Marla Fleserlu3
`
`1Department of Medicine, Pituitary Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
`2Neuroendocrinology Clinic, Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias
`Médicas y Nutricion Salvador Zubiran, Mexico City, Mexico
`5Departments of Medicine and Neurological Surgery, and Northwest Pituitary Center. Oregon Health & Science
`University, 3181 SW Sam Jackson Park Road (BTE 472), Portland, Oregon 97239, USA
`
`Correspondence
`should be addressed
`to M Fleseriu
`Email
`
`fleseriuGohsuedu
`
`Abstract
`
`JournalofEndocrinology
`
`Cushing's disease (CD) is characterized by an ACTH-producing anterior corticotrope pituitary
`adenoma. If hypothalamus—pituitary—adrenal (HPA) axis physiology is disrupted, ACTH
`secretion increases, which in turn stimulates adrenocortical steroidogenesis and cortisol
`production. Medical treatment plays an important role for patients with persistent disease
`after surgery, for those in whom surgery is not feasible, or while awaiting effects of
`
`radiation. Multiple drugs, with different mechanisms of action and variable efficacy and
`tolerability for controlling the deleterious effects of chronic glucocorticoid excess, are
`available. The molecular basis and clinical data for centrally acting drugs, adrenal
`steroidogenesis inhibitors, and glucocorticoid receptor antagonists are reviewed, as are
`potential novel molecules and future possible targets for CD treatment. Although progress
`has been made in the understanding of specific corticotrope adenoma receptor physiology
`and recent clinical studies have detected improved effects with a combined medical therapy
`approach, there is a clear need for a more efficacious and better-tolerated medical therapy
`for patients with CD. A better understanding of the molecular mechanisms in CD and of HPA
`axis physiology should advance the development of new drugs in the future.
`
`Key Words
`cortisol
`
`Cushing’s disease
`ACTH
`
`pasireotide
`mifepristone
`ketoconazole
`LCl699
`
`cabergoline
`
`VVVVVVVV
`
`Journal of Endocrinology
`
`(2014) 223. R19—R39
`
`Introduction
`
`is caused by an adrenocortico-
`(Zushing's disease ((21))
`tropin (ACTH)-secreting pituitary tumor that stimulates
`cortisol production by the adrenal glands. Morbidity and
`mortality are significantly increased if hypercortisolemia
`is left untreated. Transsphenoidal surgery, performed by
`an experienced neurosurgeon, is currently considered the
`first-line treatment. Medical treatment is commonly used
`to control the deleterious effects of persistent chronic
`glucocorticoid excess and 24-h urinary free cortisol (UFC)
`normalization still represents the gold standard to
`evaluate the efficacy of most medical treatments.
`Glucocorticoid exerts effects through the glucocorti-
`coid receptor (GR) and as the GR is expressed in almost
`
`
`http:/fioerendocrinology-iournals.org
`DOI: 10.1530/10E-14-0300
`
`© 2014 Society for Endocrinology
`Printed in Great Britain
`
`every human tissue, conditions of glucocorticoid excess,
`such as CD,
`result
`in deleterious effects on cell
`
`metabolism. Pharmacological agents can be classified as
`adrenal steroidogenesis blockers, centrally acting drugs,
`and GR antagonists. Recent
`research has evaluated
`
`chimeric compounds that work synergistically through
`membrane interaction or dimerization of both somato-
`
`statin receptors (SSTRs) and dopamine D2 receptors in
`corticotrope cells. Although no available medical agents
`surpass the efficacy of surgical therapy, new treatments
`acting directly at
`the pituitary adenoma have been
`approved for use and highlight
`the importance of
`targeting the pituitary corticotrope. A review of the
`
`‘ Published by Bioscientifica Ltd.
`
`Teva Pharmaceuticals USA, Inc. v. Corcept Therapeutics, Inc.
`PGR2019-00048
`
`Corcept Ex. 2034, Page 1
`
`

`

`L‘- cutva: ammo: and 24 ittsr nu
`
` 223 :2
`
`Treatment of Cushing’s d/sease
`
`R20
`
`)ournalofEndocrinology
`
`mechanisms of current and future medical
`
`treatment
`
`modalities for CD is provided.
`
`Overvlew of CD therapy
`
`is characterized by chronic
`Cushing’s syndrome (CS)
`overproduction of cortisol resulting in significant morbidity
`and, when left untreated, increased mortality (Newell-Price
`et al. 2006, Dekkers et a1. 2007). CS is classified as ACTH-
`
`dependent and -independent. The most common etiology
`(70—80%) of CS is CD, caused by an ACTH-secreting
`pituitary adenoma or, more rarely, by ectopic ACTH or
`corticottopin-releasing hormone (CRl-i) production that
`may result in corticotrope hyperplasia (Newell-Price et a].
`2006, Biller et ul. 2008). Chronic cortisol excess leads to a
`
`typical clinical phenotype (Table 1). Although epidemio-
`logical data on CD are limited, population-based studies
`indicate an incidence of 1.2—2.4 per million (Arnardottir &
`Sigurjonsdottir 2011, Holland et al. 2011) with a preva-
`lence of approximately 39 per million population
`(Feelders et a1. 2012). Compared with the general
`population or patients with other pituitary adenomas,
`patients with hypercortisolism have a four times higher
`
`mortality risk if untreated and cardiovascular disease
`remains the leading cause of death (Newell-Price et al.
`2006, Dekkers er a1. 2007, Feelders & Hofland 2013).
`
`line therapy
`Transsphenoidal surgery is the first
`achieving 65—90% disease remission for microadenomas
`(tumors <1 cm; Billeretal. 2008,1‘ritosetal. 201 l, Fleseriu
`2012), and lower remission rates (<65%) for macroade-
`
`nomas (tumors >1cm) (Aghi 2008). The risk of CD
`recurrence can reach 25%, 3 years after surgery (Patil
`et al. 2008). Second and third line therapies such as a
`second pituitary surgery (Patil et a1. 2008, Fleseriu 2012),
`pituitary irradiation (conventional and/or stereotactic)
`(Estrada et al. 1997, Tritos et a1. 2011) and bilateral
`
`adrenalectomy (Young 81 Thompson 2005, Assie et al.
`2007, Chow et ul. 2008, Rilzel et ul. 2013) have been
`
`utilized with variable results and specific complications.
`
`Targets for medlcal treatment of CD
`
`The hypothalamus—pituitary—adrenal (HPA) axis is orga-
`nized into three regions:
`the hypothalamus, pituitary
`gland, and adrenal glands (Fig. 1). CD is caused by a
`pituitary tumor; therefore, medical therapy should ideally
`
`'l'able 1 Clinical features and associated morbidity observed in Cushing's syndrome
`Clinical feattl'es
`
`
`
`Frequency (96)
`
`Increased weight (centripetal obesity, supraclavicular region, and upper back)
`Skin changes (round face, facial plethora, and skin atrophy)
`Decreased libido
`Menstrual irregularity
`Muscle proximal weakness
`Hirsutism
`Violaceous striae
`Easy bruising
`Associated morbidity
`Obesity
`Hypertension
`Glucose intolerance or diabetes mellitus
`Osteoporosis
`Psychiatric symptoms
`Dyslipidemia
`Increased infections and decreased wound healing
`Renal calculi
`Venous thromboembolism
`Avascular necrosis in femoral head
`Specific for Cushing's disease
`Headaches
`Visual problems (bitemporal hemianopsia)
`Other anterior pituitary hormone deficiencies
`Alterations with severe hypercortisolism
`Weight reduction (with ectopic ACTH secretion by malignancy)
`Hypoalbuminemia
`Skin hyperpigmentation
`Hypokalemia and metabolic alkalosis
`
`80-95
`80—90
`25—90
`75—80
`60—80
`70—75
`55—65
`45—65
`
`40-95
`60—80
`50—80
`40—75
`50—70
`40—70
`1 5—30
`1 5—20
`10—20
`5—1 0
`
`0—37
`0—33
`0—25
`
`10-50
`15—35
`10—15
`4—10
`
`Boscaro et al. (2001), NeweII-Price et al. (2006), Biller er al. (2008), Bertagna (2006), and Greenman (2010).
`
`
`http:/fjoeendocrinology-ioumals.org
`DOI: 10.1530'106-14‘0300
`
`© 2014 Society for Endocrinology
`Printed in Great Britain
`
`‘ Published byBioscientifica Ltd.
`
`Teva Pharmaceuticals USA, Inc. v. Corcept Therapeutics, Inc.
`PGR2019-00048
`
`Corcept Ex. 2034, Page 2
`
`

`

`:1 and :4 «List nu
`
`Treatment of Cush/ng’s dlsease
`
`223 :2
`
`R21
`
`Centrally actlng urugs
`
`
`
`
`t
`
`Corticotrope
`adenoma
`
`Serotonlnerglc antagonlsts
`Cyproheptadine
`
`GABA agonlsta
`““9”": 3"“
`
` Temozolomlde
`. Etomidate
`
`Dopamlne agonlsts
`. Cabergoline
`Chlmerlc compounds
`
`Retlnolc acid
`
`Sicrondogcncsis blocks rs
`Imldazoles
`Ketoconazole
`Fluconazole
`
`L
`
`Mlteprlstone (nuns)
`7
`7 7
`
`kMifepriStone
`
`Cell membrane
`mm of peripheral
`tissue
`k
`‘
`
`Nuclear
`membrane
`WM DNA
`
`Inhibition of cell
`transcription
`
`Figure ‘l
`The hypothaIamus—pituitary—adrenal (HPA) axis and targets of drugs used
`for treating Cushing‘s disease. Under physiological conditions, cortisol
`synthesis and production are tightly regulated by the HPA axis.
`Adrenocortlcotropln (ACTH)-producing cells in the anterior pituitary
`respond to hypothalamic corticotropin-releasing hormone (CRH) and
`arglnine vasopressln (AVP). After binding to melanocortin type 2 receptor
`(MCZR), ACTH induces the steroidogenic enzymes to increase the
`biosynthesis of cortisol and will decrease ACTH and CRH secretion. Pituitary
`corticotrope ACTH-secreting adenomas, however, function autonomously
`
`JournalofEndocrinology
`
`and overstimulate cortisol production at the adrenal cortex. The
`pharmacotherapies for ACTH-secreting pituitary corticotrope adenomas
`are categorized by the site of action into three groups: i) centrally acting
`agents or neuromodulators, which inhibit ACTH release from pituitary
`adenomas, ii) adrenal steroidogenic inhibitors, which black one or several
`steps in cortisol blosynthesls and Ill) the glucocortlcold receptor-blocking
`agent mifepris'tone. Green arrows indicate activation and red arrows/lines
`indicate inhibition.
`
`target the corticotrope cell adenoma. However, as gluco-
`corticoids represent the end hormone of the HPA axis and
`hypercortisolism induces comorbidities, steroidogenic
`inhibition was the first
`therapy used. Indications for
`medical therapy in patients with CD are summarized
`in Table 2.
`
`Adrenal steroidogenesis blockers
`
`Adrenal cortical atrophy was first documented in dogs
`treated with the insecticide dichlorodiphenyldichloro-
`ethane (DDD; Nelson 8t Woodard 1949). This observation
`
`led to the development of the use of o,p’-DDD or
`mitotane, initially for adrenal cancer and CS (Cueto «St
`Brown 1958, Southren et a1. 1961). Subsequently, ampho-
`nenone B (Hertz et al. 1956, Thorn et al. 1956),
`
`aminoglutethimide (Camacho er al. 1967), metyrapone
`(Gower 1974), trilostane (Potts et a1. 1978), and ketocona-
`
`zole (Pont et al. 1982) were identified as steroidogenic
`inhibitors (Fig. 2 and Table 3).
`
`
`http:/Iioe.endocrinology-ioumals.org
`DOI: 10.1530lJOE-14-0300
`
`© 2014 Society for Endocrinology
`Printed in Great Britain
`
`Ketoconazole
`
`Introduced as an antifungal agent, ketoconazole exerts
`endocrine side effects indicating its possible therapeutic
`efficacy in lowering cholesterol
`levels.
`Indeed,
`the
`imidazole derivative ketoconazole was noted to cause
`
`gynecomastia associated with lower plasma testosterone
`and cortisol values (Table 3; Font et at. 1982). Ketocona-
`
`zole was first used in the treatment of a patient with a
`cortisol-producing adrenal adenoma in 1983 (Engelhardt
`et al. 1983), and has been used off-label since then.
`
`Ketoconazole inhibits the side-chain cleavage complex
`(P4SOscc, CYP11A1, or 20,22 desmolase),
`llfi-hydroxyl-
`ase, and 17a—hydroxylase (Table 4) (Feldman 1986, Loli etal.
`1986). As a result of effective inhibition of cholesterol side-
`
`chain cleavage and 17-hydroxylase/17,20 lyase activity,
`ketoconazole may reduce androgen synthesis; therefore,
`effects on hirsutism are favorable (Fig. 2). There are
`inhibitory effects on several cytochrome P450 enzymes,
`mainly CYP3A4, CYP2C9, and CYP1A2 (Feldman 1986,
`Feelders et al. 2010a, Fleseriu 8r Petersenn 2012),
`
`‘ Published byBioscientifica Ltd.
`
`Teva Pharmaceuticals USA, Inc. v. Corcept Therapeutics, Inc.
`PGR2019-00048
`
`Corcept Ex. 2034, Page 3
`
`

`

`L‘- catva: ammo: and 24 “in nu
`
` 223 :2
`
`Treatment of Cushmg’s (1159558
`
`R22
`
`)ournalofEndocrinology
`
`Table 2 Use of medical
`indications and needs
`
`
`therapy in Cushing's disease —
`
`Before or after pituitary surgery
`Preparation for surgery
`Patients with contraindications for surgery
`High operation risk
`Patients unwilling to undergo surgery
`After unsuccessful surgery
`Amelioration or control of the metabolic effects of
`hypercortisolemia
`Potentially life-threatening complications
`Patients awaiting effects of pituitary radiation
`Whenever a definitive treatment is delayed
`Primary medical therapy
`Low probability of surgical cure
`Unfavorable localization
`Invisible adenomas
`Adenoma without optic chiasm compression
`
`Fleseriu et al. (2007), Biller et al. (2008), Castinetti et al. (2008), Godbout
`er al. (2010), Fleseriu (2012), and Feeiders 81 Hofland (2013).
`
`but minimal effect on aromatase enzyme (Miller &
`
`Crapo 1993).
`The first clinical reports on patients with CD described
`
`normalization of urinary or plasma cortisol values with
`ketoconazole doses of 600—1200 mg/day (Loli er al. 1986,
`
`Tabarin et al. 1991, Miller 8r Crapo 1993). Ketoconazole
`treatment is usually started at 200 mg twice a day, and
`
`is achieved at 600—1200 mg/day
`biochemical effect
`(Fable 3; Nieman 2002, Fleseriu et al. 2012). It has been
`
`suggested that ketoconazole may also have inhibitory
`effects on ACTH secretion by corticotrope tumor cells as
`AC1"H shows no significant increase despite confirmed
`reduction in cortisol levels (Loose et al. 1983, Loli et al.
`1986, Jimenez-Rema et al. 1989, Tabarin et al. 1991).
`
`Escape from pharmacological control has been reported
`(Sonino et al. 1991). Administered as a monotherapy,
`ketoconazole decreases cortisol
`levels in 30-80% of
`
`patients (Sonino et ul. 1991, Sonino & Boscaro 1999,
`Castinetti et al. 2008). Results from a recent large multi-
`
`center retrospective study (n=200) revealed normaliza-
`tion of UFC levels measured at the last follow-up in 49% of
`
`patients (Castinetti et al. 2014). Ketoconazole also blocks
`the GR at high concentrations in cultured hepatoma cells
`(Loose et al. 1983). Reportedly, hepatotoxicity (Sonino
`1987, Sugar etal. 1987, Tabarin et a1. 1991) was mild in the
`study and resolved after drug withdrawal (Castinetti et al.
`
`2014). Ketoconazole has also been used in older patients
`(> 75 years of age) with good tolerance and disease control
`(Berwaerts et al. 1998).
`
`The use of ketoconazole has been questioned after
`warnings from the European Medicine Agencies and the
`
`
`http:/fjoeendocrinology-iournals.org
`DOI: 10.1530'106-14‘0300
`
`© 2014 Society for Endocrinology
`Printed in Great Britain
`
`US Food and Drug Administration (FDA) of potential
`hepatotoxicity (Castinetti et al. 2014). Long-term safety
`remains to be prospectively studied.
`In contrast
`to
`mitotane (which causes hypercholesterolemia), ketocona-
`zole interferes with the conversion of lanosterol
`to
`
`leading to low cholesterol concentrations.
`cholesterol,
`Ketoconazole absorption requires an acidic environment,
`precluding the use of proton pump inhibitors or H2
`receptor blockers. Currently, drug availability is limited
`in many countries.
`
`Fluconazole
`
`Fluconazole appears to have similar effects to ketoco-
`nazole; however, data are limited. Fluconazole inhibits
`
`llB-hydroxylase and 17-hydroxylase activities (Fig. 2) and
`blocks cortisol production in primary cultures of human
`adrenocortical cells (van der Pas etal. 2012). Fluconazole has
`
`been shown to decrease 11-deoxycortisol production in
`H295R cells and reduce cortisol secretion in HACIS cells and
`
`primary cultures (van der Pas et al. 2012). In cultures of
`normal adrenals, fluconazole suppressed corticosterone,
`17-hydroxypregnenolone, and androstenedione levels,
`whereas concentrations of progesterone, deoxycorti-
`costerone, and 11-deoxycortisol were increased (Fig. 2).
`Fluconazole slightly increased StAR protein mRNA
`expression (van der Pas at al. 2012). Results from clinical
`studies indicated that fluconazole at adose of 100 mgtwice a
`day successfully controlled UFC levels in two patients (Riedl
`et al. 2006). Together, the results of these in vitro and in vivo
`studies indicate that fluconazole can be used to control
`
`cortisol hypersecretion in patients with CD (Table 3).
`Neither ketoconazole nor fluconazole were shown
`
`to affect the mRNA levels of steroidogenic enzymes or
`cell number: therefore; a single dose is unlikely to have a
`long-term effect (van der Pas et al. 2012).
`
`Metyrapone
`
`Metyrapone (SU-4885 : Metopirone) inhibits IIB-hydroxyl-
`ase (Fig. 2) (Liddle et al. 1959) and 17a-, 18-, and 19-
`hydroxylase (Table 4)
`(Gower 1974). Cortisol
`levels
`decrease after 2 h of the first dose (Verhelst et a1. 1991).
`
`Doses range from 500 mg to 4.5 g/day, or 6 g/day in divided
`doses (usually starting with 250 mg four times daily,
`Table 3; Nieman 2002, Fleseriu 2012). Notably, and similar
`to ketoconazole, rapid uptitration of metyrapone is
`essential to achieve a full effect (Kamenicky et al. 2011,
`Castinetti et al. 2014). The strong cortisol-lowering effect of
`metyrapone can be accompanied by the loss of negative
`
`‘ Published byBioscientifica Ltd.
`
`Teva Pharmaceuticals USA, Inc. v. Corcept Therapeutics, Inc.
`PGR2019-00048
`
`Corcept Ex. 2034, Page 4
`
`

`

`lam-.10: and xi vim w
`
`Treatment of Cushmg’s dlsease
`
`D (div-L“:
`
`223 :2
`
`R23
`
`2.
`
`22
`
`24
`
`as
`
` Cholesterol
`
`CYP1 7A1
`
`Zona tasciculata
`Glucocorticoids
`
`CYP17A1
`
`Zona reticularis
`Androgens
`
`HSD332
`
`Pregnenolone
`
`—t-
`
`Progesterone
`
`CYP11BI
`
`CYP21 A2 —t-
`
`Deoxycortioostorone
`
`cvpnaz -t :-
`
`Ooniooslerone
`
`CYP1132 - t .
`
`Aldosterone
`
`17-OH-pregnenoione
`
`- t a
`
`1 7-OH-progesterone
`
`
`
`1 1-Deoxycortisoi
`
`Zona glomerulosa
`Mineralooortlcolds
`
`JournalofEndocrinology
`
`Figure 2
`Adrenocortical steroidogenic pathways. A simplified diagram of adrenal
`steroidogenesis is depicted. Cushing's disease is almost always caused by
`a pituitary corticotrope adenoma that oversecretes corticotropin (ACI'H).
`ACTH stimulates the adrenal gland to start steroid synthesis. After
`activation of MCZR by ACTH, the StAR protein is phosphoryiated. Then,
`StAR facilitates cholesterol transport across the mitochondrial inner
`membrane. Cholesterol is the common precursor of the steroid molecules
`and, after several enzymatic reactions, is ultimately converted into
`
`biologically active aldosterone. cortisol, or androstenedione that is further
`processed to testosterone in testicles. The zona glomerulosa, fasciculata,
`and reticularis are the three adrenal cortex histological zones. which
`synthesize steroid hormones with mineraloconlcold, glucocortlcoid, or
`androgenic properties respectively. Enzyme nomenclature is given in detail
`in Table 1. CYP11A1, CYPHB1, and CYP1’lBZ are located in the
`mitochondria, and the remaining enzymes are located in the smooth
`endoplasmic reticulum.
`
`feedback and ACTH inhibition (Fig. 1). Increments in
`ACTH secretion may override steroidogenic blockade that
`may occur during the first month following initiation of
`treatment. Nevertheless, prolonged metyrapone activity
`in patients with (ID has been shown, despite a rise in
`plasma ACTH levels (Verhelst et al. 1991). ACTH also
`overstimulates production of adrenal androgen and miner-
`alocorticoid precursors (e.g. desoxycorticosterone). More-
`over, aldosterone biosynthesis is more severely affected
`than that of cortisol (Gower 1974). Therefore, several side
`
`effects related to metyrapone treatment limit its clinical use
`in patients with CD (Table 3). However, combination
`
`therapy has been proposed to improve tolerance and
`increase efficacy (Table 5; Verhelst et a1. 1991, Kamenicky
`er al. 2011). Despite safety concerns about the use of this
`drug in pregnant women, metyrapone has been used
`sporadically during pregnancy (Lindsay er al. 2005, Karaca
`et al. 2010). Additional long-term studies with metyrapone
`
`
`http:/fioe.endocrinology-ioumalsorg
`DOI: 10.1530106'14‘0300
`
`© 2014 Society for Endocrinology
`Printed in Great Britain
`
`in patients with CD are warranted. Currently, metyrapone
`has limited availability in most countries.
`
`Mitotane
`
`1 -(o-Chlorophenyi)-1-(p-chiorophenyi)-2,2—dichloroethane
`
`(o,p’-DDD) (Lysodren; mitotane), an insecticide analog of
`dichlorodiphenyltrichloroethane has been extensively used
`
`to treat all forms of hypercortisoiism (Boscaro et al. 2001).
`Mitotane’s mechanism of action was initially described in
`animal studies as lipid accumulation and atrophy of
`fasciculate and reticularis regions of the adrenal cortex. Effects
`
`on the zona glomerulosa were only observed after prolonged
`therapy (Cueto St Brown 1958). Within 12 h of treatment in
`dogs, electron microscopy revealed rupture of mitochondrial
`cristae, followed by mitochondrial swelling, lysis, and cell
`death (Miller St Crapo 1993). Conversion of cholesterol to
`pregnenolone was markedly impaired indicating that
`
`‘ Published byBioscientifica Ltd.
`
`Teva Pharmaceuticals USA, Inc. v. Corcept Therapeutics, Inc.
`PGR2019-00048
`
`Corcept Ex. 2034, Page 5
`
`

`

`lournalofEndocrinology
`
`0 (Java: ammo: and 24 (LUL nu 223 :2
`
`Treatment of Cushmg’s d/sease
`
`R24
`
`Table 3 Doses and relevant side effects of drugs used in the medical treatment for Cushing's disease
`
`
`
`
`Steroidogenesis inhibitors
`Ketoconazole
`Fluconazole
`
`Metyrapone
`
`Mitotane
`
`Etomidate
`
`LCI699
`
`Centrally acting drugs
`Cabergoline
`Bromocriptine
`Pasireotide
`
`Retinoic acid
`
`400—1200 mg/day
`200 mg/day
`
`0.5—4.5 g/day
`
`2—5 g/day
`
`Bolus 0.03 mg/kg i.v.;
`followed by
`0.1-0.3 mg/kg per h
`4—100 mg/day
`(investigational drug)
`
`0.5-7 mg/week
`1—30 mg/day
`750—2400 pig/day
`
`10-80 mg/day
`(research drug)
`
`Glucocorticoid receptor antagonist
`Mifepristone
`300—1200 mg/day
`
`
`
`
`
`Hepatitis. cholestasis, gynecomastia, gastrointestinal upset.
`edema, skin rash. Hypogonadism in men. Fluconazole may be
`better tolerated
`
`Acne, hirsutism, lethargy, dizziness, ataxia, nausea, hypertensive
`crisis, hypokalemia, edema, and adrenal insufficiency
`Gastrointestinal distress, impaired concentration and dizziness,
`gynecomastia, hepatitis, cholestasis, hyperlipidemia,
`prolongation of bleeding time, and adrenal insufficiency.
`Effect antagonized by spironolactone
`Nephrotoxicity, sedation, pain at the infusion site. anaphylactic
`reactions. coughing, hiccups, nausea, vomiting, and
`myoclonus
`Fatigue, nausea, headache, and hypokalemia
`
`Nasal congestion, nausea, postural hypotension, and headache.
`Gradual increase minimizes side effects
`
`Gastrointestinal-related conditions, hyperglycemia, cholel'rtiasis,
`nausea, abdominal pain, fatigue, headache, hypotension, and
`adrenal insufficiency
`Xerophthalmia and arthralgias
`
`Hypokalemia, worsening of hypertension, adrenal insufficiency,
`endometrial hyperplasia, fetal loss, gastrointestinal
`complaints, fatigue, nausea, headache, arthralgia, vomiting,
`and edema. Contraindicated in women planning pregnancy
`
`leffcoate et al. (1977), Robinson et al. (1987). Verhelst et al. (1991), Castinetti et al. (2009), Fleseriu et al. (2012), Heyn et al. (2012), Nieman (2002). and
`Feelders 8r Hofland (2013).
`
`side—chain cleavage of cholesterol was the major enzymatic
`
`increment from 0.5 to 1 g/day (Table 3; Luton er a1. 1979).
`
`step affected inhibiting the cytochrome P450 enzymes
`CYPllAl
`(mitochondrial side-chain cleavage enzyme),
`IIB-hydroxylation (CYPllBI), and 18-hydroxylation
`
`Results from a recent retrospective study revealed remission in
`48 (72%) out of 67 patients treated for long-term CD after a
`median of 6.7 (5.2—8.2) months (Baudry et a1. 2012) at lower
`
`(CYPllBZ), and non-P450 enzymes (SB-hydroxysteroid
`dehydrogenase) (Fig. 2; Miller & Crapo 1993). Mitotane also
`
`doses compared with adrenal cancer.
`Despite its effectiveness, mitotane therapy is compli-
`
`stimulates CYP3A4 expression, reducing cortisol bioavail-
`ability (Kroiss et al. 2011). Therefore, mitotane has three
`effects:
`i) adrenocorticolytic,
`ii) modification of steroid
`metabolism, and iii) direct inhibition of steroid biosynthesis.
`Although used to treat adrenocortical carcinoma, mitotane
`has proven effective in patients with CD (Bledsoe et al. 1964,
`Baudry er al. 2012). Mitotane displays a relatively slow onset
`
`of action compared with other steroidogenesis inhibitors
`and saturation can be expected 2—3 months after initiation
`of therapy (Luton et al. 1979, Fleseriu 8r Petersenn 2012).
`Overall, 80% of patients achieve normalization of urinary
`markers; however, 60% relapsed after therapy withdrawal
`indicating a low level of adrenolytic action (Luton eta]. 1979,
`Schteingart et a1. 1980). Sustained remission after mitotane
`
`discontinuation has been reported in 30% of patients
`measured at a mean follow-up of 37 months (Miller 6t
`Crapo 1993). Doses are approximately 4 g/day, with a gradual
`
`
`http:/fjoeendocrinology-ioumalsorg
`DOI: 10.153MOE-14‘0300
`
`© 2014 Society for Endocrinology
`Printed in Great Britain
`
`cated by several side effects (Table 3). Owing to an
`accelerated metabolism of exogenous steroids, especially
`hydrocortisone, replacement doses must be increased to
`avoid adrenal crises (Robinson et a1. 1987). Mitotane may
`
`lead to restoration of gonadal function and fertility:
`therefore, effective contraception is advisable for female
`patients (Miller 8r Crapo 1993). Mitotane is stored in
`
`adipose tissue for 2 years after administration ends and
`should not be used in women considering pregnancy
`within 5 years of discontinuation (Leiba eta]. 1989). Owing
`to the stimulatory effect of mitotane on cortisol-binding
`globulin levels (Nader er al. 2006), serum cortisol measure-
`ments are not useful and monitoring of urinary or serum
`free cortisol is the best index of response (Alexandraki et ul.
`
`2010). Replacement therapy requirements are higher than
`normal, due to interference of hormone-binding proteins
`as well. Mitotane combined with pituitary irradiation was
`
`‘ Published byBioscientifica Ltd.
`
`Teva Pharmaceuticals USA, Inc. v. Corcept Therapeutics, Inc.
`PGR2019-00048
`
`Corcept Ex. 2034, Page 6
`
`

`

`L‘- ccntvc‘: ammo: and 24 titer nu
`
`Treatment of Cushmg’s dlsease 223 :2
`
`R25
`
`
`Table 4 Enzymes that catalyze the initial reaction in the pathways of steroid hormone biosynthesis
`Nomenclature“
`
`Localization and common name
`
`Previous
`
`Current
`
`Inhibitors
`
`References
`
`Mitochondrial
`StAR protein
`Side-chain cleavage (scc)
`enzyme or 20,22 desmolase
`
`StAR
`P450scc
`
`StAR
`CYP11A1
`
`11B-hydroxylase
`
`P450 C11
`
`CYP11B1
`
`Aldosterone synthase,
`18-hydroxylase
`
`P450 C11AS
`
`CYP11BZ
`
`Imidazolesb
`Imidazoles
`Etomidate
`Mitotane
`
`Aminoglutethimide
`Imidazoles
`Mitotane
`Aminoglutethimide
`Metyrapone
`Mitotane
`LCI699
`Aminoglutethimide
`Metyrapone
`
`3B-HSD
`
`33-HSD
`
`Trilostane
`
`Walsh et al. (2000)
`Ca macho et al. (1967), Schulte et al.
`(1990), and Miller & Crapo (1993)
`
`Camacho et al. (1967), Gower (1974),
`Feldman (1986), Loli et al. (1986),
`and Miller & Crapo (1993)
`
`Camacho et al. (1967), Gower (1974),
`Miller & Crapo (1993), and Calhoun
`et al. (2011)
`
`Potts et al. (1978), Dewis et al. (1983),
`and Engelhardt & Weber (1994)
`
`Smooth endoplasmic reticulum
`17B-hydroxysteroid dehydro-
`genase, 35-hydroxysteroid
`dehydrogenase
`17a-hydroxylase/17,20 lyase
`
`Camacho et al. (1967), Gower (1974),
`Imidazoles
`Feldman (1986), Loli et al. (1986),
`Aminoglutethimide
`and Miller 8: Crapo (1993)
`Metyrapone
`Ca macho et al. (1967)
`Aminoglutethimide
`CYP21A2
`P450 C21
`21-Hydroxylase
`
`
`
`P450 aroAromatase CYP19
`Ca macho et al. (1967)
`Aminoglutethimide
`
`P450 C17
`
`CYP17
`
`)ournalofEndocrinology
`
`“P4505cc or CYP11A1, i.e. gene family 11, subfamily A, polypeptide 1.
`hImidazoles:ketoconazole, fluconazole, econzale. and/or miconazole.
`
`shown to be efficacious in non-controlled clinical trials
`
`(Luton et al. 1979, Schteingart et al. 1980).
`
`disturbance, sepsis, severe psychosis, and in preoperative
`instability (Heyn et a1. 2012, Preda et al. 2012).
`
`Etomidate
`
`LCI699
`
`The imidazole anesthetic agent etomidate (Amidate or
`Hypnomidate) was observed to decrease postoperative
`cortisol values in patients during anesthesia (Feldman
`1986). Subsequently,
`it was discovered that etomidate
`inhibits
`the
`llB-hydmxylase
`enzyme
`((ZVPl 1 Bl),
`17a—hydroxylase (CYP17A1), and the cholesterol side-chain
`cleavage complex (P4505cc, or CYP11A1, or 20,22 desmo-
`lase), thus blocking multiple steps ofsteroidogenesis (Fig. 2).
`It is the only parenteral steroidogenesis inhibitor available
`and provides rapid hypercortisolemia control (Allolio et al.
`1988, Schulte etal. 1 990). An iv. bolus injection of etomidate
`at a low non-hypnotic dose (0.03 mg/kg) followed by
`constant infusion of 0.3 mg/kg per h for 24 h (Table 3)
`decreases serum cortisol in a dose-dependent manner with
`significant suppression after the first 5 h with a maximum
`effect after 11 h (Allolio et al. 1988, Schulte er al. 1990).
`
`Glucocorticoid replacement to prevent adrenal insufficiency
`is warranted after 24 h of etomidate infusion. Clinical use of
`
`etomidate in CS has been limited by sedative side efi‘ects, but
`could be safe and effective in significant biochemical
`
`
`httpzlfjoeendocrinology-iournalsorg
`DOI: 10.153MOE-14‘0300
`
`© 2014 Society for Endocrinology
`Printed in Great Britain
`
`First characterized as an aldosterone biosynthesis inhibitor
`for primary aldosteronism and essential hypertension,
`LC1699 is an iS-hydroxylase (aldosterone synthase)
`inhibitor blocking the conversion of hydroxycortico-
`stemne tn aldosterone. It also inhibits llB-hydroxylase
`(CYPIIBI) in a similar manner to the R-enantiomer of
`
`that blocks the hydroxylation of
`fadrozole (FAD286)
`deoxycortisol
`to cortisol as well as CYPllBZ blocking
`the conversion of deoxycorticosterone to corticosterone
`(Fig. 2) (Calhoun et al. 2011). LC1699 is currently under
`investigation as a treatment for CD (Tritos et a1. 2011,
`Feelders 8t Hoflancl 2013). In a phase II proof of concept
`study of LC1699, rapid UFC normalization in 11 out of
`12 patients with CD, all achieving >50% reduction in
`baseline UFC, was observed. Doses ranged from 4 to
`100 mg/day for 10 weeks (Bertagna et al. 2014). As
`expected, ACTH increased; 45% of cases had ACTH more
`than twice that of baseline. Most adverse events were mild
`
`or moderate (Table 3). Based on these promising results,
`a larger phase II] trial is awaited.
`
`‘ Published byBioscientifica Ltd.
`
`Teva Pharmaceuticals USA, Inc. v. Corcept Therapeutics, Inc.
`PGR2019-00048
`
`Corcept Ex. 2034, Page 7
`
`

`

`L‘- cutva: Minus and 24 (List an 223 :2
`
`Treatment of Cushmg’s d/sease
`
`R26
`
`Table 5 Overview of agents that have been tested as combination drug therapy for Cushing's disease. Doses: cabergoline from 0.5
`to 3 mg/week, ketoconazole 200 to 1200 mglday, pasireotide 300 to 750 pig/day, mitotane 3 to 5 g/day, metyrapone 3 to 4.5 g/day,
`and octreotide 300 to 1500 rig/day. The studies are described in the text
`
`
`
`
`UFC normalization (%)
`
`References
`
`Compounds
`
`Patients (n)
`
`Monotherapy
`
`Combination
`
`Barbot er a]. (2014)
`Feelders et al. (2010b,c)
`
`Ketoconazole +cabergoline
`Pasireotide + cabergoline + ketoconazole
`
`14
`17
`
`Cabergoline+ketoconazole
`Vilar er al. (2010)
`—
`Total/average
`Severe hypercortisolism/aggressive tumor
`4
`Vignati & Loli (1996)
`Ketoconazole +octreotide
`1
`Bode er al. (2010)
`Temozolomide+ pasireotide
`11 CS (4 CD)
`Kamenicky et a]. (2011)
`Mitotane + metyrapone+ ketoconazole
`
`
`Total/average
`—
`16
`
`12
`43
`
`20—30
`30
`
`25
`27
`
`0
`0
`0
`0
`
`80
`50 P+C
`90 P + C + K
`70
`73
`
`0
`100
`100
`66
`
`)ournalofEndocrinology
`
`UFC, urinary free cortisol.
`
`Trilostane (Vetoryl), an inhibitor of 30-hydroxy-
`steroid, was withdrawn from human use in the USA in
`
`1994 (Potts et a]. 1978, Dewis et a]. 1983, Engelhardt 8r
`Weber 1994). Aminoglutethimide (Cytadren; Camacho
`eta]. 1967, Misbin eta]. 1976) is rarely used in the treatment
`of patients with CD. The mechanisms of action, common
`doses, and main adverse effects of aminoglutethimide are
`listed in Tables 2 and 3.
`
`Centrally acting drug treatments
`
`Human corticotrope adenomas display responsiveness to
`neurohumoral influences such as dexamethasone suppres-
`sion, lysine vasopressin, thyrotropin-releasing hormone,
`and metyrapone administration. Dexamethasone is a
`synthetic glucocorticoid extensively used to demonstrate
`the sensitivity of HPA axis negative feedback to gluco-
`corticoids for differential diagnosis of (ES. if the HPA axis
`
`is normal, any supraphysiologicai dose of dexamethasone
`is sufficient to suppress pituitary ACTH secretion. ACTH
`secretion can also be suppressed in most patients with CD
`as pituitary corticotrope adenomas are only relatively
`resistant to inhibition of glucocorticoid negative feedback.
`Hypophyseal ACTH secretion can be influenced by
`serotonin antagonists, dopamine or gamma-amino buty-
`ric acid (GABA) agonists, somatostatin receptor ligands
`(SRLs), retinoic acid, and temozolomide (Fig. 1). In
`addition to biochemical effects, CD has been associated
`with periodic hormonogenesis (cyclical CD) or spon-
`taneous remission, indicating that some cases of CD may
`be due to altered neuroregulatory influences (Van Cauter
`8r Refetoff 1985, Dickstein et a]. 1991, Colao et a]. 1997,
`Nieman 2002).
`
`
`httpzlfjoeendocrinology-ioumalsorg
`DOI: 10.153MOE-14‘0300
`
`© 201

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket