throbber
Toronto
`
`McGraw-Hill
`l‘i’lizoui‘ ii. PI‘HLHHINU Du isiioi'
`
`Tenth Edition
`
`
`
`New York
`Milan
`
`Chicago
`New Delhi
`
`San Francisco
`San Juan
`
`Lisbon
`Seoul
`
`London
`Singapore
`
`Madrid
`Sydney
`
`Mexico City
`
`DEF-ABIRA-0000465
`
`MYLAN PHARMS. INC. EXHIBIT 1111 PAGE 1
`
`

`

`MCGmw-Hiil
`A Division of The McGraw-Hill Companies
`
`32
`
`Goodman and Gilman’s Tllli Pl{ARMACOLUUlCAL HASH (ll; Trl {filial-3151l'I‘lC‘S. 10/e
`
`Copyright © 2001, 1996, 1990, 1985, 1980, 1975, 1970, 1965, 1955, 1941 by The McGraw-Hill
`Companies, Inc. All rights reserved. Printed in the United States of America Except as
`permitted trnder the United States Copyright Act of 1976, no part of this publication may be
`reproduced or distributed in any form or by any means, or stored in a data base or retrieval
`system, without the prior written permission of the publisher.
`
`1234567 890 DOWDOW 0987654321
`
`ISBN 00741354694
`
`This book was set in Times Roman by York Graphic Services, Inc. The editors were Martin I.
`Wonsiewicz and John Mr Morriss; the production supervisor was Philip Galea; and the cover
`designer was Marsha Cohen/Parallelogram. The index was prepared by Irving Condé Tullar and
`Coughlin Indexing Services, Inc.
`RR. Donnelley and Sons Company was printer and binder.
`
`This book is printed on acid—free paper.
`
`Library of Congress Cataloging-in—Publication Data
`
`Goodman and Gilman’s the pharmacological basis of therapeuticsi-IOth edr / [edited by]
`Joel G. Hardman, Lee E. Limbird, Alfred Goodman Gilman.
`p.
`; cm.
`Includes bibliographical references and index.
`ISBN 0—07-135469—7
`
`_
`
`QV 4 G6532 2002]
`
`International Edition is not available in North America.
`
`I. Title: Pharmacological basis of therapeutics.
`2. Chemotherapy.
`1. Pharmacology.
`H. Goodman, Louis Sanford
`III. Gilman, Alfred
`IV Hardman, Joel G.
`V. Limbird, Lee E.
`VI. Gilman, Alfred Goodman
`[DNLMz 1. Pharmacology,
`2. Drug Therapy.
`RM300 G644
`2001
`615/.7—d021
`
`INTERNATIONAL EDITION ISBN 0—077112432—2
`Copyright © 2001. Exclusive rights by The Mchw-Hill Companies, Inc, for manufacture and export
`This book cannot be re~exported from the country to which it is consigned by McGraW~HilL The
`
`2001030728
`
`DEF-ABIRA-0000466
`
`MYLAN PHARMS. INC. EXHIBIT 1111 PAGE 2
`
`

`

`CHAPTER
`
`ADRENOCORTICOTROPIC HORMONE;
`ADRENOCORTICA-L STEROIDS AND
`THEIR SYNTHETIC ANALOGS;
`INHIBITORS OF THE _
`v
`g SYNTHESIS AND ACTIONS OF
`ADRENOCORTICAL
`
`HORMONES
`
`1649
`
`Adrenocorticotmpic hormone (ACTH, also called corticotropin) and the steroid hormone
`products of the adrenal cortex are considered together in this chaptenbecause the ma—
`jor physiological and pharmacological ejfects of ACTH result from its action to increase
`the circulating levels of adrenocortical steroids. Synthetic derivatives of ACTH are used
`principally in the diagnostic assessment of adrenocortical fitnction. As all of the known
`therapeutic effects of ACTH can be achieved with corticosteroids, synthetic steroid hor-
`mones generally are used instead of ACTHfor therapeutic applications.
`Corticosteroids and their biologically active synthetic derivatives dtfier in their
`metabolic (glucocorticoid) and electrolyte—regulating (mineralocorticoid) activities. These
`agents are employed at physiologiCal doses for replacement therapy when endogenous
`"production is impaired. In addition, glucocorticoids are potent suppressors of inflamma-
`tion, and their use in a wide variety of inflammatory and autoimmune diseases makes
`them among the most frequently prescribed classes of drugs. Because they exert efl‘ects
`on almost every organ system, the clinical use of and withdrawal from corticosteroids
`are'complicated by a number of serious side efiects, same of which are life-threatening.
`Therefore, the decision to institute therapy with corticosteroids always requires carefitl
`consideration of the relative risks and benefits in each patient.
`Agents that inhibit various reactions in the steroidogenic pathway and thus alter the
`patterns of secretion of adrenocortical steroids are discussed, as are synthetic steroids,
`such as mtfepristone (see also Chapter 58), that inhibit glucocorticoid action. Agents
`that inhibit the action of aldosterone are presented in'Chapter 29; agents used to inhibit
`growth of steroid—dependent tumors are discussed in Chapter 52.
`
`Bernard P. Schimmer and Keith L. Parker
`
`history. The clinical importance of the adrenal glands was
`first appreciated by Addison, who described fatal outcomes
`in patients with adrenal destruction in a presentation to the
`South London Medical Society in 1849- These studies, pub-
`lished subsequently (Addison, 1855), were soon extended by
`Brown-Séquard, who demonstrated that bilateral adrenalectomy
`was fatal in laboratory animals. It later was shown that the
`adrenal cortex, rather than the medulla, was essential for sur-
`. Vival in these experiments. Further studies demonstrated that the
`adrenal cortex regulated both carbohydrate metabolism and fluid
`
`and electrolyte balance. Efforts by a number of investigators ul-
`timately led to the isolation and characterization of the various.
`adrcnocorticosteroids. Studies of the factors that regulated car—
`bohydrate metabolism (termed glucoconicoids) culminated with
`the synthesis of conisonc, the first pharmacologically effective
`glucocorticoid to be available in large amounts. Subsequently,
`Tate and colleagues isolated and characterized a distinct cor»
`ticostcroid, aldostcmnc,
`that had potent effects on fluid and
`electrolyte balance (and therefore was termed a mineralocor-
`ticoid). The isolation of distinct corticosteroids that regulated
`
`DEF-ABIRA-0000467
`
`MYLAN PHARMS. INC. EXHIBIT 1111 PAGE 3
`
`

`

`1650
`
`SECTION XII HORMONES AND HORMONE ANTAGONISTS
`
`Ser—T r-Ser-Met-
`
`roival-Lys-Va!-Tyr-Pro-Asn-Gly-Ala-Glu-
`Asp-Glu-éeréAlaéGlu-Ala-hh'e-hrmLeuéalu-Phe I
`'
`Figure 60—1. Processing ofPOMC to ACTH and the sequence
`of ACTH.
`
`A schematic overview of the pathway by which pro-opio—
`melanooortin (PQMC) is converted to ACTH and other
`peptides in the anterior pituitary is shown. The light
`blue boxes behind the ACTH structure indicate regions
`identified as important for sucroidogenic activity (residues
`6—40) and binding to the ACTH receptor (15—13). The
`amino acid sequence of human ACTH is shown. LPH,
`lipotropin; MSH, melanocyte—stimulating hormone; PC1,
`prohormone convertase l.
`x.
`
`number of other biologically important peptides, including
`endorphins, lipotropins, and the melanoeyte—stirnulating
`hormones (MSH), also are produced from the same
`prcursor.
`
`Actions on the Adrenal Certex. ACTH stimulates the
`
`adrenal cortex to Secrets glucocorticoids, mineralocorti—
`coids, and weak androgens such as androstenedione and
`dehydroepiandrosterone, which can be converted periph-
`erally into more potent androgens. Based on histological
`analyses, the adrenal cortex originally was separated into
`three zones5the zona glomerulosa, zone fasciculata, and
`20113 reticularis. Functionally, it is more useful to view the
`adrenal cortex as two discrete compartments: the outer
`zona glomerulosa,.which secretes the mineraiocorticoid‘
`aldosterone, and the inner zonae fasciculata/reticularis,
`
`which secrete the gluco'conicoid cOrtisol as well as the
`adrenal androgens (Figure 60—2).7The biochemical basis
`for these differences in steroidogenic output has been de-
`fined in considerable detail. Cells of the outer zone have
`
`receptors for angiotensin II and express aldosterone syn-
`thase, an enzyme that catalyzes the tcrminai reactions in
`mineralocorticoid biosynthesis. In contrast, cells of the in-
`ner zones lack receptors for angiotensin H and express two
`enzymes, steroid 17a-hydroxylase (P45017aland 11f}—
`hydroxylase (P450115),
`that catalyze the production of
`
`_
`
`carbohydrate metabolism or fluid and electrolyte balance ulti—
`mately led to the concept that the adrenal cortex comprises two
`largely independent units: an outer zone that produces mineralo-
`corticoids and an inner region that‘synthesizes glucocorticoids
`and weak androgens.
`Studies of the adrenocortical steroids also played a key
`part in delineating the role of the anterior pituitary» inendocrine
`function. As early as 1912, Cushing described patients with hy—
`percorticism, and later recognized that pituitary basophilism rep-
`resented the cause of the adrenal overactivity (Cushing, 1932),
`thus establishing the link between the anterior pituitary and
`adrenal function. These studies ultimately led to the purifica‘
`tion of ACTH (Astwood et (11., 1952) and the determination of
`its chemical structure. ACTH was further shown to be essential
`in maintaining the structural integrity and steroidogenic capac-
`ity of the inner cortical zones. The role of the hypothalamus in
`pituitary control was established by Harris (1948), who further
`postulated that a soluble factor produced by the hypothalamus
`activated ACTH release. These investigations culminated with
`the determination of the structure of corticotropin—releasing hor»
`mone (CRH), a hypothalamic peptide that regulates secretion of
`ACTH from the pituitary (Vale 3! (IL, 1981).
`Shortly after synthetic cortisone became available, Hench
`and colleagues demonstrated the dramatic effect of glucocorti—
`coids and ACTH in the treatment of rheumatoid arthritis (Hench
`er al., 1949). These studies set the stage for the clinical use
`of corticosteroids in a wide variety of diseases, as discussed
`below.
`
`glucocorticoids.
`
`ADRENOCORTICOTROPIC
`
`HORMONE (ACTH;
`CORTICOTROPIN)
`
`The sequence of human ACTH, a peptide of 39 amino
`acids, 'is shown in Figure 60—1. Whereas removal of a
`single amino acid at the amino terminus considerably im-
`pairs biological activity, a number of amino acids can be
`removed from the carboxy—terminal end without a marked
`effect. The structure—activity relationships of ACTH have
`been studied extensively, and it is believed that a stretch of
`four basic amino acids at positions 15 to 18 is an important
`determinant of high-affinity binding to the ACTH recep-
`tor, whereas amino acids 6 to 10 are important for receptor
`activation (Imura, 1994). As discussed in Chapter 23 and
`' as schematized in Figure 60—1, ACTH is synthesized as
`part of a-1arger precursor protein, pro-opiomelanocortin
`(POMC), and is liberated from the precursor through pro—
`teolytic cleavage at dibasic residues by the enzyme pro-
`hormone convertase 1. Impaired processing of POMC due
`to a mutation in prohormone convertase 1 has been im—
`plicated in the pathogenesis of a human disorder present-
`ing with adrenal insufficiency. lntriguingly, these patients
`also exhibit childhoodobesity, hypogonadotropic hypo~
`gonadism, and diabetes (Jackson et‘ al., 1997), suggesting
`other proteolytic targets for prohormone convertase 1. A
`
`DEF-ABIRA-0000468
`
`MYLAN PHARMS. INC. EXHIBIT 1111 PAGE 4
`
`

`

`CHAPTER 60 ACTH; ADRENOCORTICAL STEROIDS AND THEIR SYNTEETIC ANALOGS
`
`1651
`
`
`
`“Case.A_1,
`
`1EUR}‘
`
`3;‘_
`Simon
`
`Fasoicdlata/
`Reticularis
`
`Medulla '
`
`Aldosterone
`
`. P45011B
`
`13450170,} 1
`
`Cortisol,
`Androgens
`
`in the gland, it is believed that the actions of ACTH to
`increase steroid hormone production are predominantly
`mediated at the level of de novo biosynthesis. ACTH, like
`most peptide hormones,
`interacts with a specific mem»
`brane receptor. As determined by gene cloning and se-
`quencing, the human ACTH receptor is a member of the
`G protein—coupled receptor family, closely resembling in
`its structure the receptors for melanocyte-stimulating hor-
`mones (Cone and Mountjoy, 1993). ACTH acts through
`the G protein Gs to activate adenylyl cyclase and increase
`intracellular cyclic AMP content. Cyclic AMP is an oblig-
`atory second messenger for most, if not all, effects of
`ACTH on steroidogenesis. Mutations in the ACTH recep—
`tor have been associated with rare syndromes leading to
`familial resistance to ACTH (Clark and Weber, 1998).
`
`lipoid adrenal hyperplasia, a rare
`
`Temporally, the response of adrenocoru'cal cells to ACTH
`has two phases: an acute phase, which occurs within seconds to
`minutes, largely reflects an increased supply of cholesterol sub—
`strate to the steroidogenic enzymes; a chronic phase, Which-oc—
`curs over hours to days, results largely from increased transcrip-
`tion of the steroidogenic enzymes. A summary of the pathways
`of adrenal steroid biosynthesis and the structures of the major
`steroid intermediates and products of the human adrenal cor—
`tex are shown in Figure 60—3. The rate-limiting step in steroid
`hormone production is the conversion of cholesterol to preg—
`nenolone, a reaction catalyzed by the cholesterol side—chain
`cleavage enzyme, designated P450m. Most of the enzymes
`required for steroid hormone biosynthesis, including P450390,
`are members of the cytochrome P450 superfamily, a related
`group of curred-function oxidases that play important roles in
`the metabolism of xenobiotics such as drugs and environmental
`pollutants as Well as in the biosynthesis of such endogenous
`compounds as steroid hormones, vitamin D, bile acids, fatty
`acids, prostaglandins, and biogenic amines (see Chapter I). The
`rate—limiting components in this reaction regulate the mobiliza—
`tion of substrate cholesterol and its delivery to the P450”,
`located in the inner mitochondrial matrix.
`The adrenal cortex uses multiple sources of cholesterol
`to ensure an adequate supply of substrate for steroidogenesis.
`These sources include (i) circulating cholesterol and choles-
`terol esters taken up via the low—densitylipoprotein (LDL)-
`and high-density lipoprotein (HDL)~receptor pathways, (2) lib—
`eration of cholesterol from endogenous cholesterol ester stores
`via-activation of cholesterol esterase, and (3) increased de novo
`biosynthesis.
`The mechanism(s) by which ACTH stimulates the tranle-
`cation of cholesterol to the inner mitochondrial matrix are not
`Well defined. Several candidate mediators of the acute deliv—
`ery of cholesterol
`to the mitochondria have been proposed,
`including a 30,000 dalton phosphoprotein induced by ACTH
`in all primary steroidogenic tissues,
`the peripheral benzodi~
`azepine receptor, and sterol carrier protein-2. The cDNA encod—
`ing the 30,000 dalton phosphoprotein (designated the Steroid-
`ogenic Acute Regulatory Protein, or SLAR) has been cloned and
`shown to activate steroidogenesis (Stocco and Clark, 1996).
`Significantly, mutations in the gene encoding StAR are found
`in patients with congenital
`
`nanaeonnnnn
`Eifisic-"”
`asoncogenes!20
`
`2nU
`5
`
`010‘
`.2112.
`q
`59‘,
`
`'
`
`if“:
`t:
`
`Figure 60-2. The adrenal cortex contains two anatomically
`and functiomlly distinct compartments.
`The majdr functional compartments of the adrenal cor-
`tex are shown, along with .the steroidogenic enzymes that
`determine the unique profiles of corticosteroid products.
`Also shown are the predominant physiologic regulators
`of steroid production: angiotensin II (A II) and K" for
`the zona glomemlosa and ACTH for the zonae fascicu—
`lata/reticularisI
`
`In the absence of the adenohypophysis, the inner
`zones of the cortex atrophy, and the production of glu—
`cocorticoids and adrenal androgens is markedly impaired.
`Although ACTH acutely can stimulate mineralocorticoid
`production by the zona glomerulosa,
`this zone is regu-
`lated predominantly by angiotensin II and extracellular
`K+ (see Chapter 31) and does not undergo atrophy in the
`absence of ongoing stimulation by the pituitary gland. In
`the setting of persistently elevated ACTH, mineralocor—
`ticoid levels initially increase and then return to normal
`(a phenomenon termed ACTH escape).
`Persistently elevated levels of ACTH, due either to
`repeated administration of large doses of ACTH or to flex—
`cessive endogenOus ACTH production, induce hyperplasia
`and hypertrophy of the inner zones of the adrenal cortex,
`with overproduction of cortisol andadrenal androgens.
`Adrenal hyperplasia is most marked in congenital disor-
`ders of steroidogenesis, where ACTH levels are continu-
`ously elevated as a secondary response to impaired cortisol
`biosynthesis.
`
`Mechanism of Action. ACTH stimulates the synthesis
`and release of adrenocortical hormdnes. As specific mech-
`anisms for steroid hormone secretion have not been de~
`fined and since steroids do not accumulate appreciably
`
`DEF-ABIRA-0000469
`
`MYLAN PHARMS. INC. EXHIBIT 1111 PAGE 5
`
`

`

`1652
`
`
`Figure 60-3. Pathways of cortigosteroid biosynthesis.
`The steroidogenic pathways used in the biosynthcsis of the corticosteroids are shown, along with the structures of the inicrmediaies and
`products. The pathways that are unique [0 the zona glomerulosa are shown in blue, whereas those unique to the zonac fasciculaia/reticularis
`are shown in gray. P4503”, cholesterol side-chain cleavage enzyme; 3,8-HSD, 3fi~hydroxysteroid dehydrogenase; P450171“ steroid 170!-
`hydroxylase; P450“, steroid 21—hydroxyiase; P4503140, aldostemne synthasc; P450nfl, steroid IXfi—hydroxylase.
`
`H3C
`
`o P
`
`rogesterone
`
`HO
`
`Pregnenolone
`
`Sfi-HSD
`
`|C:
`
`
`
`
`
`
`
`
`
`
`DEF-ABIRA-0000470
`
`MYLAN PHARMS. INC. EXHIBIT 1111 PAGE 6
`
`

`

`CHAPTER 60 ACTH; ADRENOCOR'I‘ICAL STEROIDS AND THEIR SYNTHETIC ANALOGS
`
`Cortisol
`
`Figure 6M._ Overview of the hypotkalamic—pituitmy-adrenal
`(HPA) axis and its bidirectional communication with the im.
`mune system.
`'
`
`The complex regulatory interactions bethen the HPA
`axis and the immune/inflammatory network are shown.
`(-9 indicates a positive regulator. 9 indicates a negative
`regulator:
`IL—l,
`interleukin-1;
`IL—2,
`interleukin-2; IL-6,
`interleukin-6; TN'F-or,
`tumor necrosis factor a; CRH,
`corticotropin—releasing hormone.
`
`stimuli can override these normal negative feedback con—
`trol mechanisms, leading to marked increases in plasma
`concentrations of adrenocortical steroids.
`
`Central Nervous System. The central nervous system (CNS)
`integrates a number of different positive and negative influences
`on ACTH release (Figure 604). These signals converge on the
`CRH neurons, which are clustered largely in the parvocellular
`region of the paraventricular hypothalamic nucleus and make
`axonal connections to the median eminence of the hypothala-
`mus (see Chrousos, 1995, see also Chapter 12). Following re-
`lease into the hypophyseal plexus, CRH is transported via this
`portal system to the pituitary, where it binds to specific mem- .
`brane receptors on corticotropes. Upon CRH binding, the CRH
`receptor activates adenylyl cyclase and increases cyclic AMP
`levels within corticotropcs, ultimater increasing both ACTH
`biosynthesis and secretion. The human CRH receptor has been
`cloned and shown to resemble most closely in sequence the cal-
`citonin/vasoactive intestinal peptide/growth hormone—releasing
`hormone family of G protein—coupled receptors (Chen et al.,
`1993).
`
`Arginine Vasopressin. Arginine vasopressin (AVP) also acts
`as a secretagogue for corticotropes, significantly potentiating
`the effects of CRH. Animal studies have revealed that the po—
`tentiation of CRH action by AVP liker plays a physiologically
`significant role in the full magnitude of the stress response. AVP
`is produced in the parvocellular neurons of the paravenuicuiar
`nucleus,
`like CRH, as well as by magnocellular neurons of
`
`'
`
`congenital disorder in which adrenal cells become engorged
`with cholesterol deposits secondary to an inability to synthesize
`anysteroid hormones (Lin et aL, 1995). This finding points to
`a key role of StAR in the regulated delivery of cholesterol to
`the steroid biosynthetic pathway.
`An important component of the trephic effect of ACTH is
`the enhancement of transcription of the genes that encode the
`individual steroidogem'c enzymes, with associated increases in
`the steroidogenic capacity of the gland. Although the molecular
`mechanisms are still under investigation, it appears that a variety
`of transcriptional regulators mediate the induction of the steroid
`hydroxylases by ACTH (Parker and Schimmer, I995).
`
`In large doses, ACTH
`Extraadrenal Effects of ACTH.
`causes a number of metabolic changes in adrenalectomized
`animals, including ketosis, Iipolysis, hypoglycemia (im-
`mediately after treatment), and resistance to insulin (later
`after treatment}. Because of the large doses of ACTH
`required,
`the physiological significance of these extra-
`adrenal effects is doubtful. ACTH also improves learn—
`ing in experimental animals; this latter effect appears to
`be nonendocrine and mediated via distinct receptors in
`the central nervous system. Patients with primary adrenal
`insufficiency and persistently elevated ACTH levels clas—
`sically are hyperpigmented. This hyperpigmentation prob
`ably results from ACTH activating the MSH receptor on
`the melanocytes, perhaps a consequence of the identity of
`ACTH and MSH in the first 13 amino acids of each of
`their sequences.
`
`p
`
`the supraoptic nucleus; it is secreted into the pituitary plexus
`
`of ACTH Secretion. Hypothalamic-
`Regulation
`Pituitary—Adrenal Axis. The rates of secretion of gluco-
`corticoids are determined by fluctuations in the release of
`' ACTH by the pituitary corticolropes. These corticotropes,
`in turn, are regulated by corticotropin—releasing hormone
`(CRH), a peptide hormone released by CRH neurons of
`the endocrine hypothalamus. These three organs collec-
`tively are referred to as the hypothalamic-pituitary—adrenal
`(HPA) axis, an integrated system that maintains appro-
`'
`‘ priate levels of glucocorticoids (see Figure 60—4 for an
`overview of this axis). There are three characteristic modes
`of regulation ofthe HPA axis: diurnal rhythm in basal
`steroidogenesis, negative feedback regulation by adrenal
`corticosteroids, and marked increases in steroidogenesis
`In response to stress. The diurnal rhythm is entrained by
`higher neuronal centers in response to sleep-wake cycles,
`such that levels of ACTH peak in the early morning hours,
`causing the circulating glucocorticoid levels to peak at ap-
`proximately 8 AM. As discussed below,.ncgative feedback
`regulation occurs at multiple levels of the HPA axis and is
`the major mechanism that operates to maintain circulating
`glucocorticoid levels in the appropriate range. Stressful
`
`DEF-ABIRA-0000471
`
`MYLAN PHARMS. INC. EXHIBIT 1111 PAGE 7
`
`

`

`1654
`
`SECTION XH HORMONES AND HORMONE ANTAGONISTS
`
`forms of hypoadrenalisrn'due to hypothalamic or pituitary
`disorders. Patients with primary adrenal insufficiency haVe
`high ACTH levels because they lack normal gluco'corti—
`cold feedback inhibition, whereas patients with secondary
`adrenal insufficiency have pituitary or hypothalamic dis-
`ease resulting in low levels of ACTH. The immunoradio—
`metric ACTH assay also is useful in difierentiating be—
`tween ACTH-dcpendcnt and ACTH-independent forms of
`hypercorticism: high ACTH levels are seen when pitu-
`itary adenomas (ta, Cushing’s disease) or nonpituitary
`tumors that secrete ACTH (i.e., ectopic ACTH) underlie
`the hypercorticism, whereas very low ACTH levels are
`seen in patients with excessive glucocorticoid production
`due to primary adrenal disorders. Despite its consider-
`able strengths, one problem with the immunoradiomeuic
`ACTH assay is that its Specificity for intact ACTH can
`lead to false, low values in patients with ectopic ACTH
`secretion; these tumors often secrete aberrantly processed
`forms of ACTH that have biological activity but do not
`react in the antibody assay.
`‘
`
`from the median eminence. After binding to specific G protein—
`coupled receptors of the V“, subtype, AVP activates phospho-
`lipase C, producing diacylglycerol and 1,4,5-inositol trisphos-
`phate as messengers to release ACTH (see Chapters 2 and 12);
`in contrast to CRH, AVP apparently does not increase ACI‘H
`synthesis.
`v
`’
`Negative Iv‘ced'back~ of Glucocarticaids. Glucocorticoids inhibit
`ACTH secretion via direct and indirect actions on CRH neurons
`to decrease CRH mRNA levels and CRH release and via direct
`effects on corticotropes, The effect on CRH release may be
`mediated by specific corticosteroid receptors in the hippocam—
`pus, which are proposed to play important roles in negative
`feedback inhibition exerted by glucocorticoids. At lower corti—
`sol levels, the mineralocorticoid (type I) receptor, which has a
`higher affinity for glucocorficoids and is the predominant form
`found in the hippocampus, is the major receptor species occu-
`pied. As glucocorticoid concentrations rise, the glucocorlicoid
`(type II) receptor also becomes occupied as the capacity of
`the ruineralocorticoid receptor is exceeded. Basal activity of the
`HPA axis apparently is controlled by both classes of receptor,
`whereas feedback inhibition by glucocorticoids predominantly
`involves the glucocorticoid receptor.
`In the pituitary, glucocorticoids act through the glueocorti»
`cold receptor to inhibit the expression of POMC in corticotropes
`as well as‘the release of ACTH. These effects are both rapid
`(occurring within seconds to minutes and possibly mediated by
`glucocorticoid receptor—independent mechanisms) and delayed
`(requiring hours and involving changes in gene transcription
`mediated through the glucocorticoid receptor).
`The Stress Response. Circumstances of stress overcome neg-
`ative feedback regulation of the HPA axis, leading to a marked
`rise in the production of corticosteroids, Examples of stress sig~
`nals include injury, hemorrhage, scvcreinfection, major surgery,
`hypoglycemia, cold, pain, and fear. Although the precise mecha-
`nisms that underlie this stress response and the essential actions
`played by the glucocorticoids are not defined fully, it is clear
`that glucocorticoid secretion is vital for maintaining homeostasis
`in these stressful settings. As discussed below, complex inter-
`actions between the HPA axis and the immune system may be
`a fundamental physiological component of this stress response
`(see Sapolsky et at, 2000; Thrubull and Rivier, 1999).
`
`culating cortisol to levels greater than 18 to 20 rig/d1 indicates
`
`Therapeutic Uses and Diagnostic Applications of
`ACTH. There are anecdotal reports that selected con—
`ditions respond better to ACTH than to corticosteroids
`(e.g., multiple sclerosis), and some clinicians continue to
`advocate therapy with ACTH. Despite this, ACTH cur-
`rently has only limited utility as a therapeutic agent. Ther-
`apy with ACTH is both less predictable and less conve-
`nient than is therapy with appropriate steroids. In addition,
`ACTH stimulates mineralocorticoid and adrenal androgen”
`secretion and may therefore cause acute retention of salt
`and water as well as virilization. While ACTH and the
`corticosteroids are not pharmacologically equivalent, all
`of the known therapeutic effects of ACTH also can be
`achieved with appropriate doses of corticosteroids at a
`lesser risk of side effects.
`
`Testing the Integrity of the HPA Axis. At present, the major
`clinical use of ACTH is in testing the integrity of the HPA axis
`to identify those patients needing supplemental steroid cover—
`age in stressful situations. Other tests used to assess the HPA
`axis include the insulin tolerance test (see Chapter 56) and the
`metyrapone test (discussed later in this chapter). ACTH puri—
`fied from animal pituitary glands is available in long—lasting
`injectable gel preparations as a gelatin solution (ELF. ACHIAR
`GEL; 40 or SOTO/vial}. Cosyntropin (conmosru) is a synthetic
`peptide that corresponds to residues 1 to 24 of human ACTH.
`At the considerably supraphysiological dose of 250 ug, cosyn-
`tropin maximally stimulates adrenocortical steroidogenesis. In
`the rapid cosyntropin stimulation test, 250 pg of. cosyntropin is
`administered either intramusculme or intravenously, with cor—
`6501 measured just before administration (baseline) and 30 to
`60 minutes after cosyntropin administration. An increase in cir—
`
`Initially, ACTH levels were mea-
`Assays for ACTH.
`sured by bioassays that measured induced steroid produc-
`tion or the depletion of adrenal ascorbic acid; such assays
`have been used to standardize ACTH amounts in differ-
`ent preparations used for both diagnostic and therapeutic
`purposes. Radioirnmunoassays were developed to quan-
`titate ACTH levels in individual patients, but they were
`not always reproducible, and their sensitivity did not al-
`ways clearly differentiate between low and normal levels
`
`of the hormone. An irnrnunoradiometric assay, which re-
`liably measures ACTH levels, is now widely available.
`This assay, which uses two separate antibodies directed at
`distinct epitopes on the ACTH molecule, considerablyin—
`creases the ability to differentiate between primary hypo-
`adrenalism due to intrinsic adrenal disease and secondary
`
`DEF-ABIRA-0000472
`
`MYLAN PHARMS. INC. EXHIBIT 1111 PAGE 8
`
`

`

`a normal response (others also have included an increase of
`7 [Lg/d1 over the baseline value as a positive reSponse, although
`this is less widely accepted). In patients with pituitary or hy—
`pothalamic diseaseof recent onset or shortly after surgery for
`pituitary tumors, this standard cosyntropin stimulation test may
`be misleading, as the duration of ACTH deficiency may have
`been insufficient to cause significant adrenal atrophy with frank
`loss of steroidogenic capacity. For this latter group of patients,
`some experts advocate a “low-dose” cosyntropln stimulation
`test, in which 1 tag of cosyntropin is administered intravenously,
`with cortisol measured just before and 30 minutes after cosyn—
`tropin administration (Abdu et al., 1999). Because cosyntropin
`is not generally available in a l-rtg dose, the standard ampoule
`of cosyntropin (250 gig) is diluted to permit accurate delivery
`of the l—ptg challenge dose, with the cutoff for a normal re-
`sponse being the same as that for the standard test. Care must
`be taken to avoid adsorption of the ACTH to plastic tubing and
`to measure the plasma cortisol precisely at 30 minutes after the
`cosyntropin injection. Although some studies indicate that the
`low-dose test is more sensitive than the standard ZSO—ug test.
`others report that
`this test also may fail
`to detect secondary
`adrenal insufficiency.
`As noted above, primary and secondary adrenocortical dis-
`eases are reliably distinguished using currently available sensi-
`tive assays for ACTH. Thus, longer-course ACTH stimulation
`tests rarely are used to differentiate between these disorders.
`CRH Stimulation Test. Ovine CRH, also termed carticorelin
`(ACTHREL), is available for diagnostic testing of the HPA axis.
`In patients with documented ACTH~dependent hypercorticism,
`CRH testing may help differentiate beIWeen a pituitary source
`(i.e., Cushing’s disease) and an ectopic source of ACTH. After
`two baseline blood samples are obtained fifteen minutes apart,
`corticorelin (1 lag/kg) is administered intravenously over a 30—
`to 60—second interval, and blood samples are obtained at 15, 30,
`and 60 minutes for ACTH measurement. It is important that the
`blood samples be handled as recommended for the ACTH as
`say. At the recommended dose, CRH generally is well tolerated,
`although flushing may occur, particularly if the dose isladmin»
`istered as a bolus. Patients with Cushing‘s disease respond to
`CRH with either a norn‘ral or an exaggerated increase in ACTH,
`whereas ACTH levels do not increase in patients with ectopic
`sources of ACTH. It should be noted that’this test is not per~
`feet: ACTH levels are induced by CRH in occasional patients ‘
`with ectopic ACTH, whereas approximately 5% to 10% of pa-
`tients with Cushingjs disease fail to respond. To improve the
`diagnostic accuracy of the CRH stimulation test, some authori—
`
`Wm
`
`ORTISOL ALDOSTERONE
`Rate of secretion under
`
`optimal conditions
`
`Concentration in peripheral plasma:
`8 A.M.
`’ 4 PM.
`
`10 rug/day
`
`0.125 mg/day
`
`16 ,ng/IDO ml
`4 rag/100 ml
`
`0.01 rig/100 ml
`0.01 [Lg/100 ml
`
`CHAPTER 60' ACTH; ADRENOCORTICAL STEROIDS AND THEIR SYNTHETIC ANALOGS
`
`1655
`
`ties advocate sampling of blood from the inferior petrosal sinus
`following peripheral administration of CRH. When performed
`by a skilled neuroradiologist, this procedure may increase diag—
`nostic accuracy with a tolerable risk of complications from the
`catheterization procedure.
`-
`
`Absorption and Fate. ACTH is readily absorbed from par-
`enteral sites. The hormone rapidly disappears from the circula-
`tion following intravenous administration; in human beings, the
`half-life in plasma is about 15 minutes, primarily due to rapid
`enzymatic hydrolysis.
`
`Toxicity of ACTH. Aside from rare hypersensitivity reactions,
`the toxicity of ACTH is primarily attributable to the increased
`secretion of corticosteroids. Cosyntropin is generally less anti-
`genic than native ACTH. Moreover, ACTH isolated from animal
`pituitaries contains significant amounts of vasopressin, which
`can lead to life—threatening hyponatremia. These factors make
`cosyntropin the preferred agent for clinical use.
`
`ADRENOCORTICAL STEROIDS
`
`The adrenal cortex synthesizes two classes of steroids: the
`corticosteroids (glucocorticoids and mineralocorticoids),
`which have 21 carbon atoms, and the androgens, which
`have 19 (Figure 60—3). The actions of corticosteroids his-
`torically Were described as glucoco

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