throbber
Vol. 7. 65—78. January I998
`
`Cancer Epidemiology. Biomarkers 8: Prevention
`
`65
`
`Review
`
`Aromatase Inhibitors as Potential Cancer Chemopreventives
`
`
`
`Gary J. Kelloff,l Ronald A. Lubet, Ronald Lieberman,
`Karen Eisenhauer, Vernon E. Steele, James A. Crowell,
`Ernest T. Hawk, Charles W. Boone, and
`Caroline C. Sigman
`Chemoprevention Branch. Division of Cancer Prevention and Control,
`National Cancer Institute, Bethesda. Maryland 20852 [G. .I. K.. R. A. L.. R. L,.
`V. E. S.. J. A. C” E. T. H.. C. W. 8.]: and CCS Associates, Mountain View,
`California 94043 [K. E.. C. C. S.]
`
`Abstract
`
`Epidemiological and experimental evidence strongly
`supports a role for estrogens in the development and
`growth of breast tumors. A role for estrogen in prostate
`neoplasia has also been postulated. Therefore, one
`chemopreventive strategy for breast and prostate cancers
`is to decrease estrogen production. This can be
`accomplished by inhibiting aromatase, the enzyme that
`catalyzes the final, rate-limiting step in estrogen
`biosynthesis. The use of aromatase inhibitors is of clinical
`interest for cancer therapy, and selective, potent
`aromatase inhibitors have been developed. Several of
`these agents have demonstrated chemopreventive efficacy
`in animal models.
`The rationale for the use of aromatase inhibitors as
`chemopreventives and identification of inhibitors to serve
`as potential chemopreventive agents are the subjects of
`this review. After background information regarding
`aromatase is presented, the data for each inhibitor are
`summarized separately. The discussion focuses on those
`inhibitors that are clinically available or in clinical trials,
`including: aminoglutethimide (Cytadren), rogletimide,
`fadrozole hydrochloride, liarozole hydrochloride,
`anastrozole (Arimidex), letrozole, vorozole, formestane,
`exemestane, and atamestane. On the basis of results from
`preclinical studies, aromatase inhibitors may be
`promising agents for clinical trials in populations at high
`risk for developing estrogen-dependent cancers.
`Total suppression of aromatase may have adverse
`effects, as is evident in postmenopausal women (increased
`osteoporosis, cardiovascular disease, and urogenital
`atrophy). However, on the basis of preclinical studies of
`chemopreventive efficacy and chemotherapeutic
`applications of aromatase inhibitors showing dose-
`response efficacy, it may be possible to obtain
`chemopreventive effects without total suppression of
`aromatase and circulating estrogen levels. Suppressing
`
`Received 6/6/97; revised 9/29/97; accepted 10/15/97.
`The costs of publication of this article were defrayed in pan by the payment of
`page charges. This article must therefore be hereby marked udverlisemen! in
`accordance with 18 U.S.C. Section 1734 solely to indicate this fact.
`lTo whom requests for reprints should be addressed. at Chemoprevention
`Branch. Division of Cancer Prevention and Control. National Cancer Institute.
`Executive Plaza North. Suite 201. 6130 Executive Boulevard. Rockville, MD
`20852.
`
`local estrogen production may be an alternative strategy,
`as suggested by the discovery of a unique transcriptional
`promoter of aromatase gene expression, 1.4, in breast
`adipose tissue. The development of drugs that target this
`promoter region may be possible.
`
`Strategies in Development of Cancer Chemopreventive
`Agents
`This paper is the third in a series on strategies used by the
`Chemoprevention Branch of the National Cancer Institute to
`develop cancer Chemoprevention drugs (1—3). One chemopre-
`ventive strategy for hormone—dependent cancers is to interfere
`with the hormones that stimulate cellular proliferation in these
`tumors. Among the most important of these targets for inter-
`vention are estrogen-responsive tumors. Estrogen production
`can be decreased by inhibiting aromatase. the enzyme cata-
`lyzing the final, rate-limiting step in estrogen biosynthesis. The
`use of aromatase inhibitors is of clinical
`interest for cancer
`
`therapy, and selective. potent aromatase inhibitors have been
`developed. The rationale for use of aromatase inhibitors as
`chemopreventives and identification of inhibitors to serve as
`potential cancer chemopreventive agents are the subjects of this
`revrew.
`
`Association of Estrogen with Carcinogenesis
`Breast. Aromatase, the enzyme that catalyzes the rate—limiting
`step in estrogen formation (4), is expressed in several tissues in
`women. In premenopausal women, the granulosa cells of ovar-
`ian follicles produce the majority of circulating estrogen. pri~
`marin in the forrn of estradiol. Estrogen is also produced
`extragonadally in liver, muscle. and fat by aromatization of
`adrenal androgens. After menopause, adipose tissue is the ma-
`jor source of circulating estrogens (5). Extragonadal production
`of estrogen primarily involves aromatization of adrenal andro—
`stenedione, resulting in estrone, a weaker estrogen than estra-
`diol (6). Epidemiological evidence strongly supports a role for
`estrogens in the development and growth of breast neoplasia.
`The most consistently documented epidemiological risk factors
`for breast cancer, early age at menarche. late age at menopause.
`late age at
`first
`full-term pregnancy. and postmenopausal
`weight gain, all increase cumulative endogenous estrogen ex-
`posure (7—9). Experimental evidence also strongly favors a role
`for estrogens in the development and growth of breast cancers
`(10.
`ll). Estrogens promote the development of mammary
`cancer in rodents and exert both direct and indirect proliferative
`effects on cultured breast cancer cells (11). Induction of en-
`zymes and proteins involved in nucleic acid synthesis (e.g.,
`DNA polymerase and thymidine kinase) and oncogenes may
`account for their direct mitogenic effects. Indirect effects of
`these hormones also occur via induction of pituitary prolactin
`secretion and expression of various growth factors (e.g.. trans-
`forming growth factor a and epidermal growth factor) and
`non-growth factor peptides (e.g., plasminogen activators).
`It
`has been estimated that 30% of breast cancers are dependent on
`estrogen for their proliferation (12, 13). Although the available
`
`Downloaded from cebpaacrjournalsorg on October 9, 2015. © 1998 American Association for Cancer Research.
`
`AstraZeneca Exhibit 2026 p. 1
`InnoPharma Licensing LLC V. AstraZeneca AB IPR2017-00905
`
`

`

`66
`
`Review: Aromntase Inhibitors as Cancer Chernopreventives
`
`data are not conclusive, there is also evidence that estrogens
`may be directly genotoxic, for example, by DNA alkylation or
`oxidation, leading to free radicals, which, in turn, bind to and
`damage DNA (e.g., Ref. 14).
`The estrogen that stimulates tumor growth can be derived
`from extratumoral or tumor sources, and the relative impor-
`tance of each is controversial. Within the breast, adipose tissue
`is the major extratumoral source of aromatase, although aro-
`matase was also detected immunocytochemically in normal
`breast epithelial cells in one study (15). Aromatase activity is
`higher in adipose tissue from breast cancer patients than it is
`from those with benign breast disease (16). In breasts with
`cancer, aromatase expression (17) and activity (18) are higher
`in quadrants bearing tumors compared with those without tu-
`mors. The exact cellular localization of aromatase expression in
`breast cancer tissue is also somewhat controversial. Immuno-
`cytochemical studies have detected aromatase in breast carci-
`noma cells (15, 19) and in stromal spindle cells in breast tumors
`(20). At the present time, it appears that both adipose and breast
`tumor cells contribute to locally high estrogen production, and
`determination of their relative importance requires further
`study.
`The effects of estrogens are mediated by the ER.2 Al-
`though only a few studies have been carried out, 100% of
`precancerous atypical hyperplastic ductal breast lesions that
`have been studied express ER (21), and ER levels are higher in
`atypical ductal hyperplasia than they are in normal breast epi-
`thelium (22). Changes in ER expression during tumor progres—
`sion are not well established, but it has been reported that
`approximately 60% of breast DCISs are ER positive (23).
`These and other epidemiological and experimental data suggest
`that all breast cancers are estrogen responsive during some
`portion of their natural history. Therefore, limiting estrogen
`exposure should be a feasible chemopreventive approach. Not-
`withstanding the observation that not all ER-positive breast
`cancers are responsive to antiestrogen therapy,
`loss of ER
`during tumor progression implies that the premalignant stages
`of the disease may be more sensitive to estrogen deprivation
`than are later stages, when some tumors have lost estrogen
`responsiveness.
`The prophylactic potential of limiting estrogen exposure
`has been demonstrated in both clinical and experimental set-
`tings. Administration of the ER blocker tamoxifen to women
`with previous breast cancer significantly decreases the risk of
`developing a new cancer in the contralateral breast (24). Nu-
`merous studies have demonstrated the chemopreventive activ-
`ity of tamoxifen (e.g., Refs. 25 and 26) and other antiestrogens
`(27) in experimental breast cancer models. A disadvantage of
`many antiestrogens is their partial estrogen agonist effects.
`A second approach to achieve estrogen deprivation in-
`volves direct inhibition of estrogen biosynthesis via aromatase
`inhibitors. As described below, aromatase inhibitors are clini-
`cally available for breast cancer therapy. Support for the fea-
`sibility of preventing breast cancer by inhibiting aromatase
`comes from clinical studies in which aromatase inhibitors cause
`tumor regression in postmenopausal breast cancer patients. In
`experimental mammary cancer models, aromatase inhibitors
`
`2 The abbreviations used are: ER, estrogen receptor; DCIS. ductal carcinoma in
`situ; BPH, benign prostatic hyperplasia; AG, aminoglutethimide; 4-0HA. 4—hy—
`droxyandrost—4—ene—3.17-dione: DMBA. 7,lZ—dimemylbenz(a)anthracene: MNU.
`N—methyl-N—nitrosourea; FDA, Food and Drug Administration: CNS, central
`nervous system; PMSG. pregnant mare‘s serum gonadotropin; ACTH, adreno—
`corticotropin; FSH, follicle-stimulating hormone; LH, luteinizing hormone.
`
`induce regression of established tumors (e. g., 28, 29) and, more
`importantly for the purposes of this discussion, prevent cancer
`development (27, 30, 31).
`Prostate. Etiological and risk factors for prostate cancer in-
`clude age of >50 years, family history, high serum testoster-
`one, high-fat diet, prostatitis, and geographical background
`(prevalence being highest in the United States, Canada, and
`northwest Europe; Ref. 32). As for breast cancer, significant
`risk for prostate cancer appears to be associated with exposure
`to steroid hormones (i.e., the high serum testosterone and high
`fat consumption). Besides testosterone, estrogens have also
`been postulated to play a role in prostate cell proliferation and
`have been implicated in BPH and prostate cancer. BPH is a
`nonmalignant enlargement of the prostate due to cellular hy-
`perplasia and hypertrophy of both the epithelial and stromal
`elements of the gland, whereas prostate cancer involves the
`epithelial tissue. Studies on the role of estrogen in BPH may be
`informative about the regulation of prostate cell proliferation,
`but it should be emphasized that prostatic intraepithelial neo-
`plasia, not BPH, is the most likely precursor of prostate carci-
`noma (33, 34).
`In men, 10—25% of estrogen is synthesized locally in the
`testes, and 75—90% arises from extraglandular aromatization of
`testosterone and androstenedione (5, 35). The estrogenzandro-
`gen ratio increases with age, presumably due to greater estrogen
`synthesis, accompanied by unchanged or decreased androgen
`production. Whether the prostate is a source of estrogen is
`controversial. Some studies have reported an absence of aro—
`matase in normal prostates (36) and BPH (37). Others have
`reported aromatase activity in normal prostate and BPH (38)
`and in both BPH and prostate cancer cells (39).
`HRS have been found in normal, BPH, and cancerous
`human prostate tissue (40). Estrogens target the normal prostate
`stroma, and the involvement of estrogen and stroma-epithelial
`interaction in BPH has recently been studied (41, 42). In dogs,
`estrogens synergize with androgens and result in complex stro-
`mal and glandular hyperplasia. This effect appears to be due to
`an estrogen-mediated increase in stromal and epithelial andro-
`gen receptor levels (43), although the induction of BPH as a
`result of injury by estrogen metabolites, followed by 5a-dihy-
`drotestosterone-stimulated growth of altered prostatic cells, has
`also been postulated (44). Increased levels of estrogen have
`been found in BPH stroma compared with BPH epithelium and
`normal prostate epithelium and stroma (45). In both normal
`prostate and BPH, stromal estrogen levels increase with age,
`resulting in an increased estrogenzandrogen ratio. These results
`suggest that estrogen is involved in the development of BPH,
`but the clinical trials of aromatase inhibitors in BPH patients
`have not been encouraging (41, 46).
`Estrogen may also play a role in prostate cancer, as sug-
`gested by studies involving rat models. In the Noble rat, pros-
`tate dysplasia can be induced by simultaneous treatment with
`testosterone and estradiol for 16 weeks but not by treatment
`with testosterone or 5a-dihydrotestosterone alone (47). Long-
`term treatment of Noble rats with testosterone induces a low
`incidence of adenocarcinomas in the dorsolateral prostate (48),
`whereas treatment with testosterone and estrogen significantly
`increases carcinoma incidence and decreases tumor latency (49,
`50). The mechanism of estrogen action on the rat prostate is
`unknown. No effect of estrogen on prostate androgen receptor
`levels was observed in estradiol- and testosterone-treated
`Wistar rats (51). In Wistar rats, estrogen treatment increased
`nuclear Sat—reductase activity, although microsomal reductase
`activity was decreased (52). Treatment of Noble rats with
`
`Downloaded from cebpaacrjournalsorg on October 9, 2015. © 1998 American Association for Cancer Research.
`
`AstraZeneca Exhibit 2026 p. 2
`
`

`

`Cancer Epidemiology, Biomarkers & Prevention
`
`67
`
`9H;(:0
`
`9’“
`tr“— 0
`
`on
`
`
`
`3951130 £5
`
`0
`
`O
`
`m
`
`see
`
`__
`
`m
`
`Cholesterol
`
`Pregnenolone
`
`Progesterone
`
`Aldosterono
`
`_._._.
`
`
`
`o
`
`1
`
`,.
`
`c=o
`
`1
`
`..
`
`¢'>=o
`
`
`355550
`
`o
`
`
`
`1 7-Hydroxypregnenolone
`
`1 1-Hydroxyprogesterone
`
`Cortisol
`
`I
`
`cum
`i=0
`«"0"
`
`1
`

`
`
`mBI-ISD
`
`————>
`Aromatase
`

`
`m
`
`DHEA
`
`o
`Androst-4-ene-3,1 7-dione
`
`no
`
`Estrone
`
`EmmAthexiatians
`
`1 7-Ketosterold Reductase
`
`SCC:
`3pHSD:
`1 7BHSD:
`1 7KSR:
`
`Side-Chain Cleavage Enzyme
`30-Hydroxysteroid Dehydrogenase
`1 7B-Hydroxysteroid Dehydrogenase
`
`1$IISD
`
`17K3R
`on
`
`o
`
`1$H$D
`17K5R
`Aromatase fijéjj
`
`-——>
`
`no
`
`Testosterone
`
`Estradiol
`
`Fig. l. Steroidogenic pathway.
`
`estradiol and testosterone was shown to result in a unique DNA
`adduct in the dorsolateral prostate (the tissue where carcinomas
`originate in this model), coincident with the appearance of
`putative preneoplastic lesions, but the structure and mechanism
`of its formation are unknown (53). This adduct may play a role
`in carcinogenesis in this model. Interestingly, Spencer et al.
`(54, 55) also identified DNA adducts in normal human prostate
`and prostate tumor biopsies. The role of estrogen in human
`prostate cancer has not yet been fully elucidated. Estradiol has
`been shown to stimulate the growth of an androgen—responsive
`human prostate cancer cell line (LNCaP) and to inhibit the
`growth of an androgen nonresponsive line (PC3) in vitro (56).
`Treatment with an aromatase inhibitor resulted in pain relief in
`some patients with prostate cancer; however,
`there was no
`correlation between clinical response and estradiol levels (57).
`
`Aromatase Activity and Regulation
`Aromatase is an enzyme complex that is localized in the en—
`doplasmic reticulum and consists of a specific cytochrome
`P450 heme protein and a flavoprotein NADPH cytochrome
`P450 reductase. It catalyzes the synthesis of estradiol from
`testosterone and estrone from androstenedione. Three separate
`hydroxylation steps are catalyzed by the enzyme, which re-
`quires 3 mol of molecular oxygen for the conversion of 1 mol
`of C1.9 androgen to C18 estrogen (6). Importantly, because
`aromatization is the last step in steroid biosynthesis, selective
`inhibition of the enzyme should not disrupt production of other
`steroids such as adrenal corticoids (Fig. 1).
`
`Although the translated region of the aromatase gene is
`identical among different tissues, the untranslated regions and
`regulatory control appear to be tissue specific. At least four
`major promoter sites have been identified that respond to go-
`nadotropins, glucocorticoids, growth factors, and cytokines. A
`unique transcriptional promoter of aromatase gene expression,
`1.4, has been identified in breast adipose tissue (6, l7).
`
`Aromatase Inhibitors
`Both nonsteroidal and steroidal aromatase inhibitors have been
`developed, and the characteristics of each class have been
`reviewed (58—68). Nonsteroidal inhibitors act by binding to a
`prosthetic heme group on the enzyme. However, because this
`heme group is present on all members of the cytochrome P450
`superfamily, these inhibitors may lack specificity and, thus.
`inactivate other steroidogenic enzymes. Unlike steroidal inhib-
`itors, nonsteroidal inhibitors lack hormonal agonist or antago-
`nist activity and are more likely to be p.o. absorbed. Structures
`of the nonsteroidal inhibitors discussed below appear in Figs. 2
`(AG, rogletimide, fadrozole, and liarozole) and 3 (anastrozole,
`letrozole, and vorozole).
`Steroidal inhibitors bind either very tightly or irreversibly
`to the active site of the enzyme and are so-called “mechanism-
`based" or “suicide” inhibitors; that is, these inhibitors compete
`with androstenedione and testosterone for the active site of
`aromatase and are then converted to reactive alkylating species
`by the enzyme, which form covalent bonds at or near the active
`site, thereby irreversibly inactivating aromatase. Recovery of
`
`Downloaded from cebpaacrjournalsorg on October 9, 2015. © 1998 American Association for Cancer Research.
`
`AstraZeneca Exhibit 2026 p. 3
`
`

`

`68
`
`Review: Aromatase Inhibitors as Cancer Chemopreventives
`
` III 3 C
`
`N H 2
`
`Rogletimide
`
`N Aminoglutethirnide
`/ 3
`T
`c H
`
`N H
`
`c l
`
`N =/
`
`Liamzolc
`
`/ N
`N \//
`
`c
`"IN
`Fadrozole
`
`Fig. 2.
`
`Piper-idinedione and imidazole nonsteroidal inhibitors.
`
`c“: 0
`
`en,
`
`o
`
`on
`
`4-OHA
`
`0
`
`c u ,
`
`c n 3
`
`o
`
`c H :
`Exemestane
`
`0
`
`en,
`
`en,
`
`C":
`
`0
`
`Atarnestane
`
`0
`
`C H
`
`a
`
`H c \§c
`
`0
`
`Plomestane
`
`"A
`
`N
`I
`N\//
`
`"so
`
`I
`O u=\
`"\//u
`
`Fig. 4.
`
`Steroidal inhibitors.
`
`H36
`
`c
`//,
`
`CH:
`
`"ac
`
`Arimidex
`
`ea,
`
`c
`f"
`N.)N\
`
`Vomzolc
`
`cm
`
`"
`[mule
`
`F":
`NI;
`N
`
`Fig. 3. Traizole nonsteroidal inhibitors.
`
`enzymatic activity depends on the rate of de novo enzyme
`synthesis. The potential advantages of using suicide inhibitors
`include potent and sustained inhibitory activity, with the pos-
`sibility of intermittent dosing schedules. Disadvantages include
`unwanted hormonal agonist (particularly androgen) or antago—
`nist effects. Structures of some of the steroidal inhibitors dis-
`cussed below appear in Fig. 4 (4-OHA, exemestane, atames—
`tame, and plomestane).
`
`Representative Agents
`Data pertinent to the potential development of these aro-
`matase inhibitors as chemopreventive drugs are summarized
`in Table l.
`
`Nonsteroidal Inhibitors
`
`3—(4~aminophenyl)—3-ethyl-2,6-piper-
`AG. AG [Cytadren,
`idinedione] is structurally related to phenobarbital and was
`originally developed as an anticonvulsive. AG was the first
`aromatase inhibitor used clinically. It is available from Ciba-
`Geigy and is approved for use in breast cancer therapy. AG has
`demonstrated chemopreventive efficacy in rat models for breast
`cancer. Administration of diets containing AG decreased
`
`DMBA-induced mammary tumor incidence and multiplicity in
`Holtzman rats (30). Here, tumor incidence was decreased from
`70% to 28% and 16% with 0.05% and 0.1% AG in the diet,
`respectively. Multiplicity was decreased from 3.4 tumors/rat to
`2.3 tumors/rat and 2.0 tumors/rat, respectively. In a study
`sponsored by the Chemoprevention Branch of the National
`Cancer Institute, a diet containing 400 mg/kg AG also de-
`creased mammary tumor multiplicity from 10.6 tumors/rat to
`5.5 tumors/rat and increased latency 30 days in MNU-treated
`Sprague-Dawley rats (27). The efficacy in the latter study was
`accomplished in the presence of increased androgen activity, as
`reflected in a significantly increased body weight in rats treated
`with AG.
`
`AG is currently approved for the treatment of postmeno-
`pausal breast cancer, and objective responses in approximately
`33% of patients have been reported (58). However,
`it also
`inhibits a number of other steroidogenic cytochrome P450-
`dependent enzymes, including cholesterol side-chain cleavage
`enzymes, IIB-hydroxylase,
`l8-hydroxylase, and 21-hydr0xy-
`lase, necessitating glucocorticoid replacement therapy. Further-
`more, it has significant toxicities. especially CNS effects. Nu-
`merous efforts to optimize AG activity toward aromatase while
`reducing inhibition of other enzymes and diminishing toxic
`effects have been attempted, including elongation of the ethyl
`substituent and replacement of the ethyl group by a cycloalkalyl
`moiety (58, 65). Although these modifications improved aro—
`matase inhibitory activity, the AG derivatives are still not very
`potent compared to other compounds discussed below (58).
`Rogletimide. Rogletimide [pyridoglutethimideg 3-ethyl-3-(4—
`pyridyl)piperidine-2,6-dione] is an AG analogue that strongly
`inhibits aromatase (Ki = 1.1 MM) but is less potent than AG (Ki
`= 0.6 [.LM; Ref. 66). However, rogletimide is more specific and
`does not block cholesterol side-chain cleavage. It also has fewer
`adverse CNS effects. The administration of 50 mg/kglday ro-
`gletirnide to rats previously treated with DMBA prevented
`testosterone-induced increases in mammary tumor size (67).
`Several clinical studies in postmenopausal breast cancer
`patients have been reported. Doses of 200-1200 mg b.i.d. sig-
`nificantly suppressed estradiol levels without having effects on
`
`Downloaded from cebpaacrjournalsorg on October 9, 2015. © 1998 American Association for Cancer Research.
`
`AstraZeneca Exhibit 2026 p. 4
`
`

`

`Cancer Epidemiology, Biomarkers & Prevention
`
`69
`
`Table 1
`Aromatasc inhibitor
`
`Preclinical and clinical of aromatase inhibitors studies that are relevant to cancer chemoprevention
`Preclinical studies
`Clinical studies
`
`Potential as chemopreventive drug
`
`Nonsteroidal inhibitors
`AG [Cytadrem 3—(4—aminophenyl)-3—
`ethyl-2,6—piperidinedione]
`
`Rogletimide [Pyridoglutethimide;
`3—ethyl-3-(4—pyridyl)piperidine-2.6~
`dione]
`
`Fadrozole [CGS 16949A; 4-(5,6.7.8-
`tetrahydromidazo(1,5-a]pyridin-5~
`yl)benzonitrile]
`
`Liarozole hydrochloride [R 75 521,
`(+/— )-5»(m-chloro-a-imidazole- ] -
`ylbenzyl)benzimidazole
`monohydrochloride]
`
`Inhibition of DMBA-induced (30) and
`MNU-induced (27) mammary gland
`tumors in rats
`
`FDA approved for treatment of
`postmenopausal breast cancer
`
`Prevented testosterone-induced
`mammary tumor growth in rats (67)
`
`In rats bearing DMBA-induced
`mammary tumors. suppressed the
`appearance of new tumors (28) and
`prevented growth of established
`tumors (28. 74. 75); also prevented
`benign and malignant spontaneous
`mammary neoplasms in rats (73)
`Inhibited growth of slow growing,
`well-differentiated androgen
`dependent. as well as androgen-
`independent prostate cancers in rats
`(84, 85)
`
`Early clinical studies show suppressed
`serum estradiol levels in
`postmenopausal breast cancer
`patients (68)
`Early clinical studies show suppressed
`serum estradiol levels in
`postmenopausal breast cancer
`patients (77, 78)
`
`Clinical trials for treatment of
`advanced prostate canCer ongoing
`(l. 52. 83, 86)
`
`Anastrozole [lCI D1033; ZD1033;
`u,a,a',a'-tetramethyl-5—(lH—l,2,4—
`triazole-l-ylmethyD-lj-
`benzenediacetonitrile, Arimidex]
`
`Letrozole [CGS 20267; 4,4’-(1H—
`1.2.4-tn'azol- l .ylmethylene)bis-
`benzonitrile]
`
`Suppressed growth and inhibited new
`mammary tumor development in
`DMBA-induced rats (93. 94).
`
`(+)-Vorozole [R83842; (+)-6—((4—
`chlorophenyl)— 1H- 1 .2,4—tn'azole- l-
`ylmethyl)-1»methy1-1H-
`benzotriazole]
`
`Potent inhibition of MNU-induced (31)
`and DMBA-induced (101)
`mammary gland tumors in rats;
`suppressed growth and inhibited
`new mammary tumor development
`in DMBA~induced rats (101)
`
`ORG 33201 l(((3aR)-trans-l-[3a-
`ethyl-9—(etliylthio)-2,3.3a,4.5.6.
`hexahydro— lH—phenalen-Z-
`yl)methyl)-1H-imidazole HCl)
`Steroidal inhibitors
`Minamestane (FCE24928; 4—
`aminoandrosta- 1 ,4,6—niene-3, 1 7-
`dione)
`
`FDA approved for treatment of
`recurrent postmenopausal breast
`cancer in patients who have failed
`tamoxifen therapy (89. 90); highly
`potent suppressor of serum estradiol
`without affecting other steroids (89)
`Clinical trials for treatment of
`postmenopausal breast cancer
`ongoing (70); early clinical studies
`showed letrozole to be a potent and
`selective suppressor of serum
`estradiol levels in postmenopausal
`breast cancer patients (95—98)
`Clinical trials for treatment of
`postmenopausal breast cancer
`ongoing (29, 70, 102, 103); early
`clinical studies showed letrozole to
`be a potent and selective suppressor
`of serum estradiol levels in
`postmenopausal breast cancer
`patients
`
`Plomestane (MDL 18962; PED; 10—
`propargylestr—4—ene-3,17—dione)
`
`Prevented new mammary tumors (13])
`and caused regression of established
`mammary tumors (128. 131. 132) in
`DMBA-treated rats
`
`Limited clinical data shows that
`plomestane decreases scrum
`estrogen in normal males (133); no
`clinical data
`
`Nonspecific cytochrome P450
`inhibitor: so significant toxicities.
`especially CNS. may preclude its
`use in a prevention setting
`Less potent inhibitor than AG. but it is
`more specific for aromatase and less
`toxic; no chemoprevention studies
`currently planned
`More potent inhibitor than AC- or
`rogletimide; no chemoprevention
`studies currently planned
`
`Besides aromatase, liarozole inhibits
`the cytochrome P450 involved in
`retinoic acid metabolism (87); this
`suggests that liarozole may be
`synergistic with retinoids; no
`chemoprevention studies are
`currently planned, but there is
`interest in exploring the drug's
`chemopreventive activity. alone and
`in combination with retinoids
`Very potent inhibitor with long half-
`life (once daily dosing possible); no
`chemoprevention studies are
`currently planned
`
`More potent than its analogue
`fadrazolc (10X in animals. 100x in
`humans) (93): no chemoprevention
`studies are currently planned
`
`Potent aromatase inhibition. few side
`effects. and possibility of
`influencing estradiol levels in
`premenopausal women (29) are of
`interest for chemoprevention: Phase
`II chemoprevention studies in breast
`and prostate are in early planning
`stage
`Less potent inhibitor than fadrazole
`(123) but more specific; data are
`insufficient for evaluating
`chemopreventive potential
`
`Analogue of exemestane with less
`potency. but also it is devoid of
`androgenic activity (125. 126):
`available data are insufficient for
`evaluating its chemopreventive
`potential; further consideration will
`depend on results of studies with
`exemestane
`Plomestane no longer being developed
`by manufacturer; no
`chemoprevention studies currently
`planned
`
`serum levels of cortisol (68). Rogletimide produced dose-de-
`pendent aromatase inhibition, but even at the maximum toler-
`ated dose of 800 mg b.i.d., it was not as effective as AG (69).
`Side effects of rogletimide include those normally associated
`with estrogen deprivation: gastrointestinal
`symptoms, hot
`flushes, dizziness, and lethargy (70).
`
`Fadrozole. Fadrozole [CGS 16949A; 4-(5,6,7,8—tetrahydromi-
`dazo(l,5—a]pyridin-5-yl)benzonitrile], an imidazole, is a potent,
`competitive aromatase inhibitor being developed by Novartis
`(Basel, Switzerland). In vitro, fadrozole demonstrated an 1C5,0
`of 4.5 nM toward human placental aromatase (71). In MCF—7
`cells, fadrozole inhibited aromatase activity and prevented tes-
`
`Downloaded from cebpaacrjournalsorg on October 9, 2015. © 1998 American Association for Cancer Research.
`
`AstraZeneca Exhibit 2026 p. 5
`
`

`

`70
`
`Review: Aromatase Inhibitors as Cancer Cbemopreventives
`
`tosterone-induced MCF—7 growth (72). p.o. administration of
`0.260 mg/kg inhibited estrogen synthesis by >90% in imma-
`ture rats stimulated with PMSG (71).
`Fadrozole has also demonstrated chemopreventive activ-
`In intact Sprague-Dawley rats bearing DMBA-induced
`ity.
`mammary tumors, p.o. administration of 2.0 mg/kg/day fadro-
`zole completely suppressed the appearance of new tumors (28).
`In a 2-year study, treatment of Sprague-Dawley rats with 1.25
`mg/kg/day fadrozole completely prevented the appearance of
`benign and malignant spontaneous mammary neoplasms (73).
`At 0.25 mg/kg/day, no malignant mammary tumors and a
`decreased number of benign tumors were seen, whereas at 0.05
`mg/kg/day, the incidences of malignant and benign mammary
`tumors were decreased.
`
`Therapeutic effects of fadrozole have been demonstrated
`in preclinical models and in clinical trials. p.o. administration of
`fadrozole inhibited the growth of established DMBA—induced
`mammary tumors (28, 74, 75) in Sprague-Dawley rats. The
`combination of fadrozole and tamoxifen resulted in signifi‘
`cantly greater tumor regression than did either treatment alone
`in intact Sprague-Dawley rats (76). Several Phase I trials have
`been conducted in postmenopausal breast cancer patients, and
`a dose of 2 mg b.i.d. resulted in >90% aromatase inhibition
`(77, 78). In a multiple-dose study, p.o. administration of 0.6—
`8.0 mg b.i.d. rapidly suppressed blood and urine estrogen
`levels, but adrenal and cortical responses were also diminished
`at the highest dose levels (79). Two studies have suggested 1
`mg b.i.d. as an optimal dose (80, 81). The main side effects of
`fadrozole are nausea, hot flushes, and somnolence (82). Fadro-
`zole is currently in Phase III clinical
`trials as second—line
`endocrine therapy in postmenopausal breast cancer patients
`( 82).
`
`Liarozole Hydrochloride. Liarozole hydrochloride [R75521,
`(+/— )—5 -(m—chloro—a-imidazole- l -ylbenzyl)benzimidazole
`monohydrochloride], an imidazole derivative developed by
`the Janssen Research Foundation (Beerse, Belgium),
`is a
`potent
`inhibitor of aromatase activity in human placental
`microsomes and rat granulosa cells in vitro (83). However,
`it also inhibits other cytochrome P450 enzymes and de-
`creases androgen, progesterone, and cortisol synthesis in
`vitro. Several studies have investigated the effects of liaro-
`zole on the growth of prostate carcinomas in rats. Dietary
`administration of 80—160 mg/kg liarozole inhibited growth
`of slow-growing, well-differentiated, androgen-dependent
`Dunning-H tumors (84, 85), as well as androgen—indepen-
`dent prostate cancers (84, 85).
`Liarozole is currently in clinical trials for the treatment of
`advanced prostate cancer. In male volunteers, administration of
`a single p.o. dose of 300 mg significantly lowered plasma
`testosterone and estradiol concentrations for 24 h and blunted
`the normal cortisol response to ACTH (83). A Phase I dose-
`escalation trial involving 38 honnone-refractory prostate cancer
`patients treated with 37.5—300 mg b.i.d. has been conducted.
`Four patients had a >50% decrease in prostate-specific antigen
`levels. In patients with measurable soft-tissue disease, two had
`partial responses, as judged by a >50% decrease in at least one
`measurable lesion (86). There was no evidence of adrenal
`insufficiency. Side effects included lethargy, somnolence, and
`body rash. All patients enrolled in this study were under max-
`imum androgen suppression (orchiectomy or gondadotropin-
`releasing hormone therapy).
`in which liarozole was
`Results from rat model studies,
`effective in androgen-independent tumor models, and from the
`Phase I trial, in which responses were seen in androgen-sup-
`
`pressed patients, have led to the suggestion that some of liaro—
`zole’s effects are due to modulation of retinoic acid metabo-
`lism. Liarozole has been shown to inhibit cytochrome P450
`isozymes that are responsible for retinoic acid catabolism (87),
`and, in fact, liarozole has been shown to increase endogenous
`levels of retinoic acid in a number of target tissues. In view of
`this, it is not surprising that liarozole exerts retinoid-mimetic
`effects in vivo (88). At the present time, the specific mecha-
`nisms responsible for liarozole's inhibition of prostate tumor
`growth are unknown and may involve inhibition of aromatase,
`retinoic acid catabolism, and, in some cases, androgen biosyn—
`thesis.
`
`Anastrozole. Anastrozole [ICI D1033; ZD1033; a,a,a’,a'-
`tetramethyl-S-(lH-1,2,4-triazole-l-ylmethyl)—1,3-benzenedi-
`acetonitrile, Arimidex]
`is a triazole developed by Zeneca
`(Wilmington, DE), which has been approved by the FDA for
`treatment of recurrent pos

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