throbber
personaluseonly.
`
`
`
`
`
`CancerInvestDownloadedfrominformahealthcarecombyJHUJohnHopkinsUniversityon12/05/14For
`
`
`
`
`
`
`
`Cancer Investigation, 16(6), 385—390 (1998)
`
`NEW DRUG
`
`Anastrozole (Arimidex®), a New
`Aromatase Inhibitor for Advanced
`
`Breast Cancer: Mechanism of Action
`
`and Role in Management
`
`Gabriel N. Hortobagyi, M.D., and Aman U. Buzdar, M.D.,
`F.A.C.P.
`
`Department of Breast Medical Oncology
`The University of Texas M. D. Anderson Cancer Center
`Houston, Texas
`
`INTRODUCTION
`
`Breast cancer is the most common cancer in American
`
`women, and is estimated to be the cause of 46,000 deaths
`per year (1—3). The incidence of breast cancer increases
`with age, with breast cancer more common in
`menopausal and postmenopausal women than in younger
`women (4). Approximately one-third of all breast cancers
`are hormone dependent; because estrogen is the primary
`steroidal mitogen, hormonal manipulation has proven to
`be a successful modality in these cases (3,5,6). Tumors
`likely to respond to endocrine therapy can be identified
`through the presence of estrogen and progesterone recep-
`tors, with positive response rates as high as 60% in
`patients with estrogen receptor (ER)-positive tumors and
`75% in cases in which both estrogen and progesterone
`receptors have been detected (6,7). Postmenopausal
`women are more likely than younger women to have
`breast tumors positive for both estrogen and progesterone
`receptors; in addition, the absolute concentration of these
`receptors may also be higher in tumors in post—
`menopausal women (8). A relatively new strategy for the
`blockade of estrogen synthesis in the management of
`
`advanced breast cancer in postmenopausal women is aro—
`matase inhibition with the selective, nonsteroidal
`inhibitor anastrozole (Arimidex®) (9—15). Aromatase
`inhibition presents a number of advantages over other
`endocrine therapies, including a well-defined mechanism
`of action, specific inhibition of estrogen synthesis, a lack
`of estrogenic effects, and lack of cross-resistance with
`antiestrogens (5). When compared with other aromatase
`inhibitors (e.g., aminoglutethimide, formestane, fadro-
`zole), anastrozole has a number of advantages, including
`high potency, high specificity for the aromatase enzyme,
`a favorable safety profile, a convenient mode of admin-
`istration, and no requirement for corticosteroid replace-
`ment therapy (9—12,15,16).
`
`ESTROGEN SYNTHESIS AND
`AROMATASE INHIBITION
`
`Steroid hormones (aldosterone, cortisol, androgens, and
`estrogens) are synthesized via a well-defined series of
`complex reactions involving cytochrome P450 hydrolases,
`lyases, and/or aromatase, with cholesterol as a precursor
`
`385
`
`Copyright © 1998 by Marcel Dekker, Inc.
`
`www.dekker.com
`
`fit it a H ‘I' n L.
`
`l w
`
`13‘?
`
`AstraZeneca Exhibit 2012 p. 1
`InnoPharma Licensing LLC v. AstraZeneca AB IPR2017-00904
`Fresenius-Kabi USA LLC v. AstraZeneca AB IPR2017-01910
`
`

`

`personaluseonly.
`
`
`
`
`
`CancerInvestDownloadedfrominformahealthcareeombyJHUJohnHopkinsUniversityon12/05/14For
`
`
`
`
`
`
`
`386
`
`Cholesterol
`
`[(cyt r450 20,12 lyase)
`l
`Pug-lanolin:e
`l
`Progeslins
`
`l
`l (cylP450 HIS/18hydroxylase)
`ldcytP45021 hydroxylase)
`.
`,
`.
`_
`Glucocortlcoids
`MJneralomrtiemds
`
`“I
`‘L(Cyt P450 17a hydroxylasc/
`17 201),“)
`Andmgens
`F“)
`
`Testosterone
`
`Androstened'lole
`
`Anastmmle —)
`
`Jr
`Elf“)!!!
`
`(cyt p450 aromatase)
`(——)
`
`i
`Estradinl
`
`Figure 1. Estrogen synthesis (16).
`
`(16,17) (Fig. 1). The aromatase enzyme acts at the last
`step in the estrogen—synthesis pathway, catalyzing the con-
`version of androgens to estrogens (16,17). Thus, inhibition
`of the aromatase reaction does not impair the synthesis
`of progestins, glucocorticoids, mineralcorticoids, or andro—
`
`Hortobagyi and Buzdar
`
`gens (3,16,17).
`Aromatization of adrenal androgen precursors in
`peripheral tissues is the primary source of estrogen in
`postmenopausal women (5,16,17). Approximately 50—100
`ug of estrone can be produced daily by the extraglandu-
`lar conversion of androstenedione, and some of this
`
`estrone is converted to produce 10—20 pg/mL of circulat-
`ing estradiol (5). In addition to its presence in adipose,
`muscle, ovarian, brain, and liver tissue, aromatase activ-
`
`ity (5—100 pg/g tissue/hr) has also been detected in breast
`tumors (5,16,18,19). Although this tumor aromatase activ—
`ity would appear to be too low to account for a significant
`amount of estradiol, biochemical measurements might
`underestimate local levels, since aromatase may be local-
`ized in specific tumor-cell types (e.g., stromal or fat cells)
`(5,16,20).
`Aromatase inhibitors can be classified as either mech—
`
`anism—based (“suicida ”) or competitive inhibitors (3,16).
`Mechanism-based aromatase inhibitors (e.g., formes—
`tane) are enzyme-specific, steroidal compounds that act
`as substrate analogues, covalently binding to the active
`site of the aromatase enzyme complex and irreversibly
`inactivating it (3,5,16,17). Competitive aromatase
`inhibitors bind reversibly to the aromatase enzyme com-
`
`Results of Preclinical Studies Evaluating the Selectivity ofAnastrozole“
`
`Table 1
`
`
`
` P-450 enzyme Species Results
`
`
`
`Rat
`20,22-Desmolase
`(cholesterol side-chain Monkey
`cleavage)
`11—Hydroxylase
`(glucocorticoid
`production)
`
`Monkey
`Dog
`
`18-Hydroxylase
`(aldosterone synthesis)
`
`Rat
`
`17—Hydroxylase/
`1 7 ,20—Desmolase
`(androgen production)
`
`Rat
`Monkey
`Dog
`
`No adrenal hypertrophy or histological changes at 100
`times the MED.b
`
`Margin of selectivity was 30—fold in monkeys, IOU-fold in
`dogs. No increase in circulating concentrations of 11-
`deoxycorticosterone, no hypokalemia, no adrenal
`hypertrophy after 6—10 mg/kg doses.
`2 ZOO-fold margin of selectivity. No effect on plasma
`aldosterone concentration, no effect on sodium or
`potassium excretion in saline-loaded animals given 10—20
`mg/kg doses.
`No effect on prostate-gland weight or plasma
`concentrations of testosterone or luteinizing hormone in
`rats given 1—10 mg/kg/day for 7—21 days. Increased plasma
`testosterone concentrations in monkeys (52x) and dogs
`(9x); no inhibition of androgen production of 10—100
`times the MED.
`
`Lanosterol— 14-
`
`Rat
`
`2 30-fold margin of selectivity. No reduction in plasma
`cholesterol following administration of 25 mg/kg in
`Dog
`demethylase
`rats or 3 mg/kg in dogs.
`(cholesterol synthesis)
`aFrom reference 15.
`bMED, maximum effective dose for aromatase inhibition.
`
`Fl:
`
`II a H 'I' E L.
`
`1
`
`5".“ Mi}
`
`AstraZeneca Exhibit 2012 p. 2
`
`

`

`personaluseonly.
`
`
`
`
`
`CancerInvestDownloadedfrominformahealthcarecombyJHUJohnHopkinsUniversityon12/05/14For
`
`
`
`
`
`
`
`Anastrozole: Role in Advanced Breast Cancer
`
`387
`
`plex and inhibit aromatization only for as long as they
`occupy the active site (3,5). Steroidal competitive aro—
`matase inhibitors (e. g., 60t—bromo-androstenedione) are
`inherently specific but are associated with agonist and
`antagonist effects on estrogen, glucocorticoid, andro—
`gen, and progesterone receptors (5,16). Compared with
`steroidal inhibitors, nonsteroidal competitive inhibitors
`(e.g., aminoglutethimide) have a greater potential for
`blockade of several cytochrome P450—mediated steroidal
`hydroxylations, but tend not to exhibit steroidal agonist
`or antagonist activity (5,16).
`The prototype aromatase inhibitor, aminoglutethimide,
`was initially developed as an anticonvulsant. This nons—
`teroidal competitive aromatase inhibitor provides
`95%—98% inhibition of aromatase; however, aminog—
`lutethimide also inhibits cholesterol side—chain cleavage,
`producing a “medical adrenalectomy” that necessitates
`corticosteroid replacement therapy (3,5,16). In addition,
`aminoglutethimide has low potency and is associated with
`a high incidence of side effects (35%) and discontinuation
`of therapy (5%) (5,16).
`In recent years, the development strategy for aromatase
`inhibitors in the management of advanced breast cancer
`has been to synthesize agents with increased potency and
`selectivity, as well as an improved toxicity profile com—
`pared with aminoglutethimide. The third-generation,
`nonsteroidal, competitive aromatase inhibitor anastrozole
`was recently approved by the U.S. Food and Drug
`Administration (FDA) for the treatment of advanced
`breast cancer in postmenopausal women following failure
`with tamoxifen therapy.
`
`ANASTROZOLE
`
`Potency and Specificity
`
`Anastrozole {2,2’-[5—(1H—1,2,4-triazol—1—ylmethyl)-
`1,3—phenylene] bis (2—methyl-propionitrile)} has a high
`intrinsic potency and is able to inhibit human placental
`aromatase activity in vitro by 50% at a concentration
`of 0.043 ug/mL (15 nM) (9,11,15). In vivo studies of
`aromatase activity in rats have shown that an oral 0.1
`mg/kg dose of anastrozole inhibits ovulation when
`administered to mature females on day 2 or 3 of the
`estrous cycle, and inhibits androstenedione-induCed
`uterine hypertrophy when administered to prepubertal
`animals for 3 days (9,11,15). Daily oral anastrozole
`doses 2 0.1 mg/kg have been shown to inhibit periph-
`eral aromatase activity in male pig-tailed monkeys,
`resulting in 50%—60% reductions in circulating estradiol
`levels (9,15).
`
`The high selectivity of anastrozole for aromatase has
`been demonstrated in a series of studies evaluating its effect
`on other cytochrome P450 enzymes involved in steroido-
`genesis in rat, dog, and monkey models (Table 1) (9,1 1,15).
`At concentrations up to 2200 times that of its maximally
`effective aromatase-inhibitory dose, anastrozole did not pro-
`duce pathological changes in the adrenal gland nor affect
`aldosterone concentrations, sodium or potassium excretion,
`or testosterone synthesis (9,11,15). In comparison, fadro—
`zole (another aromatase inhibitor) has been shown to
`increase 1 1—deoxycorticosterone concentrations in monkeys,
`and to reduce aldosterone concentrations, as well as alter
`
`Table 2
`
`Estrogen Suppression by Anastrozole”
`
`
` Population Dose regimen Results
`
`
`
`29 Male volunteers
`(6-7/dose)
`
`0—60 mg,
`single dose escalation
`
`Doses 2 7.5 mg produced at least an 80% suppression of estradiol.
`
`14 Healthy
`postmenopausal
`female volunteers
`
`8 Healthy
`postmenopausal
`female volunteers
`
`7 days; placebo
`
`0.5 or 1 mg daily
`for 14 days
`
`Estradiol lowered to limits of detection of assay by 3—4 days of treatment in
`2/6 patients in 0.5 mg group and 7/7 patients in 1 mg group. Suppression
`maintained for at least 6 days after study drug discontinued.
`
`3—mg single dose, Estradiol levels lowered 70% (first dose) to 80% (subsequent dosing)
`followed 3 days later
`compared with placebo. Suppression maintained for 4 days after last dose.
`by 3 mg daily for
`Circulating levels of estrone minimally suppressed by first dose,
`7 days; placebo
`significantly decreased with subsequent dosing.
`crossover
`
`l9 Postmenopausal
`5 mg/day for first Estradiol concentration suppressed 80% from baseline to limits of detection of
`women with advanced 14 days, followed by
`assay. Estrone lowered 69%—86%; estrone sulfate lowered 83%—92%.
`breast cancer
`10 mg/day for an
`
`additional 14 days
`
`aFrom references 9712, 15.
`
`Fl:
`
`51 a H 'I' E L.
`
`1
`
`r.“ M}
`
`AstraZeneca Exhibit 2012 p. 3
`
`

`

`personaluseonly.
`
`
`
`
`
`CancerInvestDownloadedfrominformahealthcarecombyJHUJohnHopkinsUniversityon12/05/14For
`
`
`
`
`
`
`
`388
`
`Hortobagyi and Buzdar
`
`Table 3
`
`Tumor Response in Two Phase III Clinical Trials Involving 263 Patients Given Arimidex®
`(1 mg daily) and 253 Patients Given Megestrol Acetate (40 mg q. id. )‘1
`
`Number Responseb Stable Diseasec
`
`Progression
`
`Prog Timed
`
`Trial #1
`
`Arimidex®
`Megestrol acetate
`Trial #2
`
`Arimidex®
`Megestrol acetate
`3From references 12—14.
`
`128
`128
`
`135
`125
`
`10.2%
`5.5%
`
`10.4%
`10.4%
`
`26.6%
`29.7%
`
`23.7%
`22.4%
`
`48.4%
`51.6%
`
`58.5%
`56.0%
`
`170 days
`151 days
`
`132 days
`120 days
`
`bEither a complete or partial objective tumor response.
`CStable for 2 6 months.
`
`dMedian time to progression; conservative estimate based on strict UICC criteria.
`
`sodium and potassium excretion in rats at doses only 5 times
`its aromatase-inhibitory dose (9,11).
`Anastrozole was not associated with direct progesto—
`genic, estrogenic, or androgenic activity in rats, even at up
`to 10 times its maximally effective aromatase-inhibitory
`dose (11,12). In addition, in rats, mice, dogs, and cats,
`anastrozole did not show any significant pharmacological
`activity other
`than aromatase inhibition (11,15).
`Anastrozole did not affect autonomic, neuromuscular, or
`
`respiratory function at an intravenous dose of 1 mg/kg.
`At an oral dose of 10 mg/kg, anastrozole did not affect the
`central nervous system (CNS), cardiovascular function
`(other than causing a small reduction in blood pressure
`and shortening of the electrocardiographic Q—T interval in
`dogs), renal function, gastrointestinal motility, gastric acid
`secretion, pain perception, inflammatory response, clotting
`ability, or local anesthetic activity.
`Anastrozole's high selectivity and lack of adverse effect
`on steroidogenesis were also demonstrated in clinical
`pharmacology studies (9—12,15).
`In healthy post-
`menopausal volunteers and patients with advanced breast
`cancer, anastrozole did not affect cortisol or aldosterone
`secretion, response to adrenocorticotrophic hormone
`(ACTH), or thyroid-stimulating hormone (TSH) concen—
`trations when administered at up to 10 mg daily (i.e., up
`to 10 times the recommended daily dose).
`
`Estrogen Suppression
`
`Clinically significant suppression of serum estradiol
`(by 80% of its pretreatment baseline value) following
`0.5—10 mg daily oral doses of anastrozole was demon-
`strated in four clinical pharmacological studies using a
`highly sensitive, validated radioimmunoassay method with
`a 3.7 pmol/L detection limit (Table 2) (9—12,15). In these
`
`studies, only about one third of postmenopausal volun-
`teers and patients who were given 0.5 mg anastrozole
`achieved estrogen suppression close to the assay limit of
`detection, whereas 100% of those given 1 mg, 75% of
`those given 3 mg, 95% of those given 5 mg, and 100% of
`those given 10 mg achieved estrogen suppression to the
`lower limit of the assay (9,11,15). Therefore, a 1 mg daily
`dose of anastrozole is considered to be the minimal dose
`
`that provides maximal estrogen suppression (Table 2).
`In a recent pharmacological study involving post—
`menopausal women with breast cancer who were given 1
`mg anastrozole daily for 28 days, whole-body aromatase
`activity was reduced by 96% (21). Plasma concentrations
`of estradiol and estrone were reduced by approximately
`85%, from 19.3 pmol/L at pretreatment to 3.0 pmol/L, and
`from 72.0 pmol/L to 11.1 pmol/L, respectively. In addition,
`plasma concentrations of estrone sulfate were reduced from
`426.2 pmol/L to 32.5 pmol/L (92% reduction).
`
`Clinical Trials
`
`In two phase III, multicenter, randomized, parallel-group
`trials, postmenopausal women with advanced breast cancer
`whose disease had progressed following antiestrogen
`(tamoxifen) therapy were given either anastrozole (1
`mg/day) or the progestin megestrol acetate (40 mg q.i.d.),
`and were evaluated with respect to objective response and
`time to disease progression (Table 3), as well as duration of
`response,
`survival,
`and quality of
`life
`(12—14).
`Approximately one-third of the patients given either anas-
`trozole or megestrol acetate derived clinical benefit from
`this therapy (i.e., a positive objective tumor response or sta-
`bilization of disease for at least 6 months) (Table 3). There
`were no statistically significant differences between the treat-
`ment groups with respect to tumor response or time to pro-
`
`fltfl H‘I’E L.
`
`1
`
`1".“ its}
`
`AstraZeneca Exhibit 2012 p. 4
`
`

`

`personaluseonly
`
`
`
`
`
`CancerInvestDownloadedfrominformahealthcarecomby.THUJohnHopkinsUniversityon12/05/14For
`
`
`
`
`
`
`
`Anastrozole: Role in Advanced Breast Cancer
`
`Table 4
`
`Role in Clinical Practice
`
`Comparison of the Incidence of Potentially Drug-Related“
`Adverse Events in Two Phase III Clinical Trials Involving 262
`Patients Given Arimidex® (1 mg daily) and 253 Patients
`Given Megestrol Acetate (40 mg q. id. )b
`Arimidex® Megestrol acetate
`
`Withdrawn due to AE
`Gastrointestinal disturbance
`Hot flushes
`Edema
`Thromboembolic disease
`
`2.7%
`29.4%
`12.6%
`7.3%
`3.4%
`
`4.0%
`21.3%
`13.8%
`13.8%
`4.7%
`
`0.8%
`1.9%
`Vaginal dryness
`11.9%c
`1.5%
`Weight gain
`34.4%C
`12.6%
`25% weight gain
`
`210% weight gain 10.7%C 2.3%
`
`
`“Anticipated to be potentially causally related to one or both therapies
`based on drug pharmacology.
`bFrom references 12—14.
`CSignificantly different from Arimidex treatment group at p S 0.01.
`
`gression, indicating that anastrozole (1 mg once daily) was
`as effective as the standard regimen of megestrol acetate
`(40 mg q.i.d.) in these patients.
`Objective tumor response, based on conservative inter—
`pretation of the stringent Union Internationale Contre 1e
`Cancer (UICC) criteria, was observed in approximately 10%
`of patients given anastrozole and 8% of those given mege-
`strol acetate. Although these rates appear low, they are con—
`sistent with those reported in other studies of second-line
`hormonal treatment using UICC criteria in patients in whom
`tamoxifen therapy failed (22—26). In addition, the observed
`response rates were not unexpected, since 36% of the
`patients in these two phase III studies had had previous cyto-
`toxic therapy, 46% had visceral lesions, 43% relapsed while
`receiving adjuvant tamoxifen, 62% had bone metastases,
`and 28% had nonmeasurable disease and therefore could
`
`not attain a partial-response (PR) classification.
`Anastrozole was well-tolerated in both clinical trials,
`with the incidence of withdrawal because of adverse
`
`events and the type, severity, and incidence of individual
`adverse events generally comparable with the results
`found with megestrol acetate. In contrast, the number of
`patients gaining weight, as well as the amount of weight
`gained, was significantly greater with megestrol acetate
`than with anastrozole (p S 0.01); also, more patients
`receiving megestrol acetate continued to gain weight over
`time (Table 4). Although some benefit may be realized in
`patients with cachexia, persistent, continuing weight gain
`is undesirable, psychologically distressing, and may be
`problematic for patients undergoing chronic treatment of
`breast cancer.
`
`Postmenopausal women with estrogen receptor-positive,
`metastatic breast cancer and who do not have rapidly pro-
`gressing visceral disease or tumor-related lung or liver dys-
`function are suitable candidates for endocrine therapy. The
`nonsteroidal antiestrogen tamoxifen is currently the most
`widely used first-line hormonal agent in patients with
`metastatic breast cancer (6,27). However, up to 53% of
`patients who initially respond to tamoxifen eventually
`exhibit tamoxifen-resistant disease and require treatment
`with an alternative endocrine agent (27). For these patients,
`the third—generation aromatase inhibitor anastrozole offers a
`new option for second-line hormonal therapy.
`Cross-resistance from tamoxifen, to anastrozole has
`not been observed, and clinical experience supports the
`safety and efficacy of anastrozole in the management of
`advanced breast cancer in postmenopausal women with
`disease progression following treatment with tamoxifen
`(12—14). Aromatase-inhibitory doses of anastrozole do
`not affect the cytochrome P450 hydroxylase or lyase
`enzymes involved in steroidogenesis, and corticosteroid
`replacement therapy is therefore unnecessary in patients
`treated with anastrozole (12). In addition, alterations in
`TSH concentrations have not been observed with anas—
`
`trozole (12). Anastrozole can be given orally in a conve—
`nient, once-daily dosing regimen; is well-tolerated; and
`is not associated with weight gain (12—14). A potential
`
`Postmenopausal patients with
`
`primary disease
`
`Postmenopausal patients with
`
`advanced or recurrent disease
`
`Adjuvant tamoxifen therapy
`
`Adjuvant tamoxifen therapy
`
`Relapse during
`
`Treatment 2-5 years
`
`Relapse
`
`therapy
`
`Anastrozole
`
`Relapse 6-12 months
`
`Aliastrmle
`
`afier discontinuing
`tamoxifen
`
`l
`
`Tamoxifen rechallenge
`
`l
`
`Relapse
`
`l
`Anastroznle
`
`Figure 2. Potential algorithm for hormonal
`metastatic breast cancer.
`
`treatment of
`
`Fl:
`
`II a H 'I' E L.
`
`l
`
`r.“ M}
`
`AstraZeneca Exhibit 2012 p. 5
`
`

`

`personaluseonly.
`
`
`
`
`
`CancerInvestDownloadedfrominformahealthcarecombyJHUJohnHopkinsUniversityon12/05/14For
`
`
`
`
`
`
`
`390
`
`algorithm for hormonal treatment of metastatic breast can—
`cer in postmenopausal women is shown in Figure 2.
`
`CONCLUSIONS
`
`Anastrozole provides a new therapeutic option for post-
`menopausal patients with advanced breast cancer whose dis-
`ease has progressed after tamoxifen therapy. Anastrozole
`offers greater selectivity and potency, improved tolerability,
`and a more convenient method of administration than do
`
`other endocrine agents. Because of its similar eflicacy, once-
`daily dosing regimen, and lack of associated weight gain,
`anastrozole would be preferred to progestins such as mege—
`strol acetate for second-line therapy in the management of
`advanced breast cancer in postmenopausal women.
`
`Address correspondence and reprint requests to: Gabriel N. Hortobagyi,
`M.D., MD. Anderson Cancer Center, 1515 Holcombe Blvd, Box 56,
`Houston, TX 77030.
`
`REFERENCES
`
`3.
`
`1. Boring CC, Squires TS, Tong T: Cancer statistics, 1992. Cancer
`42:19—38, 1992.
`2. National Center for Health Statistics: Vital Statistics of the United
`States, 1987, Vol 2, Mortality, Washington DC, US. Government
`Printing Office, 1990.
`Ibrahim NK, Buzdar AU: Aromatase inhibitors: Current status.
`Am J Clin Oncol 18:407—417, 1995.
`4. Glass A, Hoover RN: Changing incidence of breast cancer. J Natl
`Cancer Inst 80:1076—1077, 1988.
`5. Santen RJ: Clinical use of aromatase inhibitors in human breast
`carcinoma. Steroid Biochem Mol Biol 402247—253, 1991.
`6. Hanis JR, Morrow M, Bonadonna G: Cancer of the breast. In:
`Cancer: Principles and Practice of Oncology, 4th ed, edited by VT
`DeVita Jr., S Hellman, SA Rosenberg, Philadelphia, PA, JB
`Lippincott, 1993. pp 1264—1332.
`7. McGuire WL: An update on estrogen and progesterone receptors
`in prognosis for primary and advanced breast cancer.
`In:
`Hormones and Cancer, Vol 15, edited by S Iacobelli, New York,
`Raven Press, 1980, pp 337-344.
`8. Bland KI, Fuchs A, WitfliSS JL: Menopausal status as a factor in
`the distribution of estrogen and progestin receptors in breast can—
`cer. Surg Forum 32:410—412, 1981.
`9. Plourde PV, Dyroff M, Dowsett M, et a1: ArimidexTM: A new oral,
`once—a-day aromatase inhibitor. J Steroid Biochem Mol Biol
`53:175—179. 1995.
`10. Yates RA, Dowsett M, Fisher GV, et a1: Arimidex (ZD1033): a
`selective, potent inhibitor of aromatase in postmenopausal female
`volunteers. Br J Cancer 73:543—548, 1996.
`
`11.
`
`12.
`13.
`
`14.
`
`15.
`
`16.
`
`17.
`
`18.
`
`19.
`
`20.
`
`21.
`
`22.
`
`23..
`
`24.
`
`25.
`
`26.
`
`27.
`
`Hortobagyi and Buzdar
`
`Walton PL, Yates RA, Dukes M: Arimidex: an overview of a new
`selective nonsteroidal aromatase inhibitor. In: Sex Hormones and
`Antihormones in Endocrine Dependent Pathology: Basic Clinical
`Aspects, edited by M Motta, M Serio, pp 311—316, 1994.
`Package Insert. Arirnidex® (anastrozole) tablets. 1995.
`Buzdar A, Jonat W, Howell A, et al: Anastrozole, a potent and
`selective aromatase inhibitor, versus megestrol acetate in post-
`menopausal women with advanced breast cancer: Results of an
`overview analysis of two phase III
`trials.
`J Clin Oncol
`14:2000—2011, 1996.
`Jonat W, Howell A, Blomqvist C, et al: A randomised trial com—
`paring two doses of the new selective aromatase inhibitor anas-
`trozole (Arimidex) with megestrol acetate in postmenopausal
`patients with advanced breast cancer. Eur J Cancer 32A:404—412,
`1996.
`
`Plourde PV, Dyroff M, Dukes M: Arimidex®z A potent and selec—
`tive fourth—generation aromatase inhibitor. Breast Cancer Res
`Treat 30: 103—1 1 1, 1994.
`Goss PE. Gwyn KMEH: Current perspectives on aromatase
`inhibitors in breast cancer. J Clin Oncol 12:2460—2470, 1994.
`Brodie AMH: Aromatase, its inhibitors and their use in breast
`cancer treatment. Pharmacol Ther 60:501—515, 1993.
`Santen RJ, Leszcynski D, Tilson-Mallet N, et al: Enzymatic con-
`trol of estrogen production in human breast cancer: Relative sig-
`nificance of aromatase versus sulfatase pathways. Ann NY Acad
`Sci 464:126—137, 1986.
`O’Neill JS, Miller WR: Aromatase activity in breast adipose tis-
`sue from women with benign and malignant disease. Br J Cancer
`56:601—604, 1987.
`Bradlow HL: A reassessment of the role of breast tumor aromati-
`zation. Cancer Res 42:3382s—3386s, 1982.
`Geisler J, Lonning P, Dowsett M, et al: A randomized, double—
`blind, multicentre, cross-over trial to evaluate in vivo inhibition of
`aromatase by Arirnidex (ZD1033) (1 mg and 5 mg p.0. o.d.) in
`postmenopausal women with breast cancer. Nottingham Breast
`Cancer Meeting—Zeneca Symposium, Nottingham, UK. 1995.
`Robertson J, Williams M, Todd J: Factors predicting the response
`of patients with advanced breast cancer endocrine. Eur J Cancer
`Clin Oncol 23:469—475, 1989.
`Muss HB, Case LD, Capizzi RL, et al: High- versus standard—
`dose megestrol acetate in women with advanced breast cancer: A
`phase IH trial of the Piedmont Oncology Association. J Clin
`Oncol 8:1797—1805, 1990.
`Mercer PM, Ebbs SR, Fraser SC, et al: Trial of amino-
`glutethirnide vs hydrocortisone as second-line hormone treatment
`of advanced breast cancer. Eur J Surg Oncol 19:254—258, 1983.
`Pronzato P, Rubagotti A, Amoroso D, et al: Second-line hor—
`monotherapy for breast cancer. Uselessness of first—line continu-
`ation. AmJ Clin Oncol 162522—525, 1993.
`Congdon J, Green S, O’Sullivan J, et a1: Megestrol acetate and
`aminoglutethirnide/hydrocortisone in sequence or in combination
`as second-line endocrine therapy of estrogen receptor positive
`metastatic breast cancer. Proc Am Soc Clin Oncol 10:43, 1991.
`Manni A: Pharmacologic manipulation of steroid hormones.
`Endocrinol Metab Clin North Am 20:825—844, 1991.
`
`Ft:
`
`II a H 'I' E L.
`
`1
`
`r.“ M}
`
`AstraZeneca Exhibit 2012 p. 6
`
`

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