throbber
3954 Vol. 10, 3954 –3964, June 15, 2004
`
`Clinical Cancer Research
`
`Report from the FDA
`
`Approval Summary for Bortezomib for Injection in the Treatment
`of Multiple Myeloma
`
`Peter F. Bross, Robert Kane, Ann T. Farrell,
`Sophia Abraham, Kimberly Benson,
`Margaret E. Brower, Sean Bradley,
`Jogarao V. Gobburu, Anwar Goheer,
`Shwu-Luan Lee, John Leighton, Cheng Yi Liang,
`Richard T. Lostritto, William D. McGuinn,
`David E. Morse, Atiqur Rahman,
`Lilliam A. Rosario, S. Leigh Verbois,
`Grant Williams, Yong-Cheng Wang, and
`Richard Pazdur
`Division of Oncology Drug Products, Center for Drug Evaluation and
`Research, United States Food and Drug Administration, Rockville,
`Maryland
`
`ABSTRACT
`Purpose: Multiple myeloma is a malignant plasma cell
`disorder accounting for about 10% of hematological malig-
`nancies. Despite treatment advances, including hematopoi-
`etic stem-cell transplantation to facilitate administration of
`high-dose cytotoxic chemotherapy, the median survival re-
`mains approximately 3 years and long-term remissions are
`rare. Bortezomib (Velcade, formerly known as PS-341; Mil-
`lennium Pharmaceuticals, Cambridge MA) is a dipeptide
`boronic acid that inhibits the 20S proteasome involved in the
`degradation of intracellular proteins, including those affect-
`ing cell cycle regulation in mammalian cells. Described
`herein are the analyses by the United States Food and Drug
`Administration (FDA) of clinical and nonclinical data sub-
`mitted in the New Drug Application. Chemistry manufac-
`turing and controls, animal toxicology, and biopharmaceu-
`tical data are described. The results of Phase I and Phase II
`clinical studies in patients with multiple myeloma are sum-
`marized. The marketing approval and postmarketing com-
`mitments are discussed.
`Results: Toxicology studies in the rat and monkey iden-
`tified hematological, lymphoid, cardiac, renal, gastrointesti-
`nal, and neurological toxicities of bortezomib. A steep dose-
`toxicity effect was noted at doses >0.9 mg/m2. Administration
`of doses >3.0 mg/m2 to monkeys resulted in cardiovascular
`collapse and death 12–14 h postdose. Histopathological evi-
`dence of axonal and myelin degeneration of dorsal root ganglia,
`peripheral nerves, and spinal cord were observed in monkeys
`and rodents; concurrent clinical observations included tremors
`and decreased activity.
`
`Received 12/22/03; revised 3/3/04; accepted 3/8/04.
`Requests for reprints: Peter F. Bross, United States Food and Drug
`Administration, HFM 755, 1401 Rockville Pike, Rockville, MD 20852.
`E-mail: bross@cber.fda.gov.
`
`Pharmacokinetic studies in patients with advanced ma-
`lignancies demonstrated that the mean elimination half-life
`after the first bortezomib dose varied from 9 to 15 h at doses
`ranging from 1.45 to 2.00 mg/m2. The drug is metabolized by
`cytochrome P450 –3A4, -2D6, -2C19, -2C9, and -1A2. Three
`Phase I studies were performed in a total of 123 patients with
`advanced malignancies. Dose-limiting toxicity included diar-
`rhea and sensory neurotoxicity. No dose-limiting hematological
`toxicity was reported.
`Safety and efficacy were evaluated in an open-label,
`Phase II study of 202 patients with multiple myeloma who
`had received at least two prior therapies and had demon-
`strated disease progression on their most recent therapy. A
`smaller dose finding study of 54 patients provided additional
`supportive information. Bortezomib was administered by
`i.v. bolus on days 1, 4, 8, and 11 in a 21-day cycle for up to
`eight cycles. The initial dose was 1.3 mg/m2 except for 28
`patients in the dose-finding study who received a 1.0 mg/m2
`dose. The primary study end point in this single-arm trial
`was response rate, easily measured and thought to correlate
`with clinical benefit in patients with myeloma. One hundred
`eighty-eight patients who met the inclusion criteria were
`included in the FDA efficacy analysis population. Complete
`responses (CRs) were observed in 5 patients and partial
`responses (PRs) in 47 patients for an overall response (OR)
`rate (OR ⴝ CR ⴙ PR) of 28%. The dose finding study of 54
`patients showed a higher response rate for patients given 1.3
`mg/m2 compared with 1.0 mg/m2 twice weekly for two of the
`3-week schedule, but the study was too small for statistical
`dose-response comparisons. The most commonly reported
`adverse events were asthenic conditions (including fatigue,
`malaise, and weakness) in 65%, nausea (64%), diarrhea
`(51%), appetite decreased (including anorexia; 43%), con-
`stipation (43%), thrombocytopenia (43%), peripheral neu-
`ropathy (37%, including peripheral sensory neuropathy and
`peripheral neuropathy aggravated), pyrexia (36%), vomit-
`ing (36%), and anemia (32%).
`Conclusions: The FDA granted marketing approval to
`Millennium Pharmaceuticals on May 13, 2003 for bort-
`ezomib for use as a single agent for the treatment of multiple
`myeloma in patients who have received at least two prior
`therapies and have demonstrated disease progression on the
`last therapy. Accelerated approval was based on a surrogate
`end point of response rate rather than clinical benefit, such
`as an improvement in survival. The recommended dose of
`bortezomib is 1.3 mg/m2 administered twice weekly for 2
`weeks (days 1, 4, 8, and 11) followed by a 10-day rest period
`(days 12–21). Accelerated approval was based on the results
`of two Phase II studies in a total of 256 patients and addi-
`tional Phase I safety information. Mandated Phase IV study
`commitments to characterize clinical efficacy and safety
`more precisely are discussed.
`
`Downloaded from
`
`on September 23, 2015. © 2004 American Association for
`clincancerres.aacrjournals.org
`Cancer Research.
`
`CFAD v. Anacor, IPR2015-01776
`ANACOR EX. 2012 - 1/12
`
`

`
`INTRODUCTION
`Multiple myeloma (MM) is a malignant plasma cell disor-
`der accounting for ⬃10% of hematological malignancies. There
`are approximately 45,000 people in the United States living with
`multiple myeloma and an estimated 14,600 new cases of mul-
`tiple myeloma are diagnosed each year.1 The reported incidence
`is 5 per 100,000 with a peak at age ⬃70 years; rates are higher
`in African Americans and in men (1). Multiple myeloma was
`first described in 1844; and in 1962, Bergsagel et al. (2) reported
`that melphalan, the phenylalanine derivative of nitrogen mus-
`tard, could induce remissions in about one-third of patients.
`Many cytotoxic regimens induce remissions, but effects on
`survival have been difficult to demonstrate despite increasing
`doses of conventional cytotoxic chemotherapy (3). Median
`overall survival does not exceed 3 years with conventional
`chemotherapy (4).
`High-dose chemotherapy followed by hematopoietic stem
`cell rescue has been shown to increase the percentage of com-
`plete remissions to almost 50% in selected patients (versus
`1–13% with conventional dose therapy), but the disease com-
`monly recurs (5, 6). High-dose chemotherapy may increase the
`CR rate and time-to-progression; however, myeloablative ther-
`apy has not consistently shown a survival improvement (7).
`Double autologous (tandem) transplantation has recently been
`shown to improve long-term survival
`in eligible patients
`less than 60 years old, but the majority of patients eventu-
`ally relapsed even after the double transplant (8). Subsequent
`treatment responses occur less frequently and are of shorter
`duration (9).
`Salmon et al. (10) first reported the efficacy of high-dose
`prednisone in this disease in 1967, and glucocorticoids are still
`a mainstay of myeloma therapy. Recent research has focused on
`other alternatives to cytotoxic chemotherapy. In 1999, Singhal
`et al. (11) reported durable responses with thalidomide in mul-
`tiple myeloma, and subsequent studies have confirmed its ac-
`tivity (12–13). In 2003, Richardson et al. (14) reported on the
`efficacy results of bortezomib, an inhibitor of the 20S protea-
`some, in advanced multiple myeloma. This article describes the
`analysis of clinical and nonclinical data that led to accelerated
`marketing approval of bortezomib for the treatment of multiple
`myeloma.
`
`RESPONSE CRITERIA IN MULTIPLE
`MYELOMA
`Multiple myeloma is characterized by the clonal prolifer-
`ation of plasma cells. Except in 1–2% of patients with nonse-
`cretory myeloma, an abnormal monoclonal immunoglobulin
`heavy- and/or light-chain paraprotein, known as M protein or M
`component, is readily quantifiable in the serum and/or urine of
`patients with multiple myeloma and has been used to measure
`the response to therapy and progression. In 1968 and 1973, the
`Chronic Leukemia and Myeloma Task Force of the National
`Cancer Institute published guidelines for the determination of
`
`1 Source of information, national program of cancer registries. Internet
`address: http://www.cdc.gov/cancer/npcr/uscs/report/.
`
`Clinical Cancer Research
`
`3955
`
`response in multiple myeloma, specifying a response parameter
`of 50% reduction in paraprotein measured by protein electro-
`phoresis (PEP) of serum (SPEP) or urine (UPEP; Ref. 15). The
`Southwest Oncology Group (SWOG) subsequently refined the
`remission criteria to require a 75% reduction in serum and a
`90% reduction in urine paraprotein (16, 17).
`CRs were rarely reported with earlier treatment options;
`however, the development of newer combination and dose-
`intensive therapy led to new proposals for assessing treatment
`response. In 1989, Gore et al. reported their response evaluation
`in a series of patients with myeloma treated with combination
`chemotherapy followed by high dose-melphalan and stem cell
`rescue (18). The Gore study reported a complete remission rate
`of 50% based on disappearance of M protein by PEP with the
`additional requirement of a confirmatory repeat electrophoresis
`finding 3 months later. Complete resolution of myeloma protein
`by PEP subsequently became a criterion for complete remission
`in the era of high-dose chemotherapy and stem-cell transplan-
`tation (7, 19 –21).
`In 1998, the European Group for Blood and Marrow Trans-
`plant (EBMT) proposed even stricter criteria for the assessment
`of CR in myeloma patients after high-dose therapy (22). These
`criteria include the complete absence of myeloma protein by
`immunofixation (IF) techniques as well as by PEP, and results
`must be confirmed at least 6 weeks later. In addition, bone
`marrow plasmacytosis must be reduced to less than 5%. Ab-
`sence of serum and urine paraprotein measured by IF has
`recently been used to define CRs for both conventional and
`high-dose regimens (23, 24, 12). Lahuerta et al. (25) published
`a retrospective study suggesting that complete remission by
`immunofixation electrophoresis status is a more sensitive pre-
`dictor of survival and time to progression than complete remis-
`sion by PEP. Differences among response categories are sum-
`marized in Table 1.
`
`THE PROTEASOME PATHWAY
`The ubiquitin-proteasome pathway is thought to play a
`critical role in the degradation of proteins involved in cell cycle
`control and tumor growth. A complex enzyme cascade first
`marks proteins destined for degradation by the covalent addition
`
`Table 1 Response criteria used in the efficacy analysis
`
`Reduction of M
`protein required
`
`SPEP
`
`UPEP
`
`IFa
`
`Response category
`EBMT
`CRb
`100%
`100%
`Negative
`90%
`50%
`NR
`PR
`50%
`25%
`NR
`MR
`SWOG remissionc
`90%
`75%
`NR
`a IF, immunofixation; M protein, myeloma protein; SPEP, serum
`protein electrophoresis; UPEP, urine protein electrophoresis; EBMT,
`European Group for Blood and Marrow Transplant; CR, complete
`response; PR, partial response; NR, not required; MR, minimum re-
`sponse; SWOG, Southwest Oncology Group.
`b EBMT CR also required ⬍5% plasma cells in bone marrow, after
`Blade et al. (22).
`c SWOG remission, after Alexanian et al. (16) and Salmon et al. (17).
`
`Downloaded from on September 23, 2015. © 2004 American Association forclincancerres.aacrjournals.org
`
`
`
`Cancer Research.
`
`ANACOR EX. 2012 - 2/12
`
`

`
`3956 Bortezomib in Multiple Myeloma
`
`of multiple molecules of ubiquitin (26). The proteasome hydro-
`lyzes only those proteins that have been marked for destruction
`by this ubiquitin enzyme cascade (27, 28). The 20S proteasome,
`the core component of the proteasome complex, is composed of
`four subunits forming a hollow cylinder that has multiple pro-
`teolytic sites on the interior wall (29). The proteasome-complex
`degrades proteins as it moves them through this cylinder. This
`20S proteasome must first bind to various other large proteins
`known as activators (PA700 and PA28 are examples) to form
`proteasome-activator complexes before it can hydrolyze the
`ubiquitin-bound protein substrates. These activators can bind to
`form different complexes, each with different protein substrate
`specificity. The most frequently studied proteasome complex is
`the 26S proteasome, a large molecule heterotrimer formed by
`the 20S proteasome and two PA700 activators (30 –33). The
`proteasome can affect cell division through ubiquination and
`degradation of inhibitory proteins through the regulation of
`nuclear transcription factors (34 –38). Evidence suggests that the
`inhibition of the proteasome can act through multiple mecha-
`nisms leading to an arrest of cell growth. Inhibition of the
`proteasome may also have other consequences. Limited in vitro
`research suggests that the inhibition of the proteasome pathway
`might lead to the accumulation of abnormal proteins, including
`prion-related protein (PrP), as demonstrated in transfected neu-
`ronal cell lines (39). The clinical consequences of this protein
`accumulation are unknown.
`
`BORTEZOMIB (PS-341, VELCADE)
`The search for molecules that could inhibit the 20S pro-
`teasome in vitro led to the discovery of bortezomib (Velcade,
`PS-341; Millennium Pharmaceuticals, Inc., Cambridge, MA), a
`small, dipeptide boronic acid that reversibly inhibits the chy-
`motrypsin-like proteolytic activity site of the 20S proteasome of
`mammalian cells (40). Bortezomib exhibits cytotoxic, growth-
`inhibitory, and antitumor activities in several in vitro and in vivo
`assay systems and binds to the proteasome at lower concentra-
`tions than it does to other tested proteases. In replicating cells in
`vitro, bortezomib appears to cause cell cycle arrest at the tran-
`sition of G2-M, and the inhibited cells then initiate apoptosis
`(41). In the standard National Cancer Institute panel of 60
`human cell lines, bortezomib inhibited cell growth and, in some
`cases, was cytotoxic for human tumor cells. The average IC50 of
`bortezomib across the 60 cell lines was 3.8 nM. In athymic mice
`implanted with both the HT-29 human colon and the PC-3
`human prostate-tumor xenograft models, bortezomib given i.v.
`weekly for 4 weeks (3 mg/m2) decreased tumor volume by up to
`50 and 65%, respectively. Resistance to bortezomib cytotoxicity
`has been noted over time in vitro. This resistance is probably not
`mediated by overexpression of
`transmembrane molecular
`pumps, such as the multidrug resistance protein.
`After in vivo dosing, inhibition of proteasome activity,
`measured in lysate from whole blood from animals or humans,
`recovers to normal in about 48 –72 h (42). Repeat dosing causes
`greater inhibition compared with a single dose at the same level
`(about 30% after a single dose compared with almost 99% after
`seven daily doses). Inhibition could be detected in tissue from
`colon, muscle, prostate, and liver but not in the testes or brain of
`rodents. The inhibition in the liver was significantly greater than
`
`in WBCs. Thus far, there is no evidence of a relationship
`between ex vivo measurements of proteasome inhibition and
`clinical efficacy.
`Bortezomib inhibited the degradation of cytochrome
`P450 –2E1 by proteasomes after ethanol induction, thus prevent-
`ing the return of intracellular expression of the enzyme to
`constitutive levels (43). Other cytochromes P450 may also be
`degraded by proteasomes after induction (44). Bortezomib has
`the potential to modify the metabolism of a broad range of
`chemicals by changing the intracellular concentration of cyto-
`chrome P-450 (45). Thus, proteasome inhibition by bortezomib
`may modify a patient’s exposure to drugs that are metabolized
`by cytochrome P-450.
`
`CHEMISTRY
`Bortezomib is a modified dipeptide boronic acid. The drug
`substance exists in its cyclic anhydride form in the solid state as
`a trimeric boroxine. The product is provided as a mannitol
`boronic ester that, in reconstituted form, exists in equilibrium
`with its hydrolysis product, the monomeric boronic acid. The
`chemical name for the monomeric form is [(1R)-3-methyl-1-
`{[(2S)-1-oxo-3-phenyl-2-[(pyrazinylcarbonyl)amino]propyl]-
`amino}butyl]boronic acid. The molecular weight is 384.24, and the
`molecular formula is C19H25BN4O4. The solubility of bortezomib,
`as the monomeric boronic acid, in water is 3.3–3.8 mg/ml in a pH
`range of 2–6.5. Bortezomib is available for i.v. injection as a sterile
`lyophilized powder in single-dose vials containing 3.5 mg bort-
`ezomib and 35 mg mannitol, USP. In this form, the drug product is
`stable and can be stored at controlled room temperature. The
`lyophilized powder drug product is reconstituted with 0.9% NaCl
`to a final concentration of 1 mg/1 ml before injection. The chemical
`structure is shown in Fig. 1.
`
`TOXICOLOGY
`Traditional
`toxicological and toxicokinetic parameters,
`neuropathological evaluations, and proteasome activity determi-
`nations were examined. Bortezomib was administered to rats as
`a single dose and twice weekly for 2 weeks and for 26 weeks.
`Bortezomib was poorly tolerated when administrated daily, even
`at very low doses. Nonclinical tolerability studies suggested that
`intermittent dosing permitted more prolonged dosing regimens
`by allowing a return toward baseline of 20S proteasome activity
`before the subsequent dose. In the 9-cycle 26-week rat study,
`
`Fig. 1 Bortezomib structure. Bortezomib is a modified dipeptide bo-
`ronic acid derived from leucine and phenylalanine.
`
`Downloaded from on September 23, 2015. © 2004 American Association forclincancerres.aacrjournals.org
`
`
`
`Cancer Research.
`
`ANACOR EX. 2012 - 3/12
`
`

`
`Clinical Cancer Research
`
`3957
`
`drug-related mortality was observed at ⱖ0.9 mg/m2 (days 23–
`180) and was due primarily to hematopoietic and lymphoid
`system depletion, along with gastrointestinal hyperplasia and
`necrosis. Histopathological changes were observed in the heart,
`liver, lung, kidney, sciatic nerve (necrosis), and spinal cord; in
`general, similar findings albeit with less severity were observed
`in scheduled-death animals. Animals who were dosed at ⱖ0.9
`mg/m2 and who survived to week 26 (end of treatment) exhib-
`ited various forms of neurotoxicity including degeneration of
`dorsal root ganglia, peripheral nerves, and spinal cord. Nephro-
`toxicity, including eosinophilic casts, inflammation, hypertro-
`phy, tubular dilation, and glomerulonephritis was observed at 26
`weeks of treatment at doses ⱖ0.6 mg/m2; a comparable inci-
`dence of tubular dilation was observed after 8 weeks of recovery
`in males. Cardiac histopathological changes included increased
`incidence of perivascular necrosis, myocardial degeneration,
`hemorrhage, and inflammation. Thrombocytopenia was ob-
`served at all dose levels. After the 8-week recovery interval,
`myocardial inflammation, cardiac necrosis, and tubular dilation
`of the kidney (males only) persisted. There appeared to be some
`indication of reversibility of other findings at this time.
`Bortezomib was administered to monkeys as a single dose,
`a daily dose for 13 days, twice-weekly for 2 weeks, and twice-
`weekly for 4- and 13-three week cycles. In the 13-cycle monkey
`study, bortezomib-related mortality was observed at dosages
`ⱖ0.9 mg/m2. Findings included severe anemia, dehydration,
`gastrointestinal diffuse mucosal hyperplasia, thrombocytopenia,
`neurotoxicity, and cardiotoxicity. There was an increased inci-
`dence of clinically observable findings, typically associated with
`neurotoxicity, in treated animals when compared with controls
`during bortezomib administration. The frequency of histopatho-
`logical findings demonstrating neurotoxicity was reduced after 8
`weeks of recovery.
`Clinical neurotoxicity was reported in monkeys, rats, and
`mice; findings included nerve degeneration of dorsal root gan-
`glia, peripheral nerves, dorsal spinal roots and dorsal tracts of
`the spinal cord at ⱖ0.6 mg/m2 (one-half of the recommended
`clinical dose of 1.3 mg/m2). Histopathological incidence of
`neurotoxic effects in monkeys appeared greater compared with
`that in rodents. Clinical observations included tremors and re-
`duced activity in monkeys; rats also exhibited reduced activity.
`Nephrotoxicity was observed at doses ⱖ0.9 g/m2 in monkeys;
`males appeared to be more susceptible. Lymphoid atrophy
`and/or necrosis occurred in thymus, spleen, lymph nodes, and
`gut-associated lymphoid tissue. In addition, necrosis, atrophy,
`and hyperplasia of the gastrointestinal tract were observed in
`monkeys surviving to 38 weeks.
`Dose- and schedule-dependent changes in AUC (area under
`the curve) and Cmax (maximum concentration) occurred in both
`species. Drug exposure with increasing dose was more linear in
`monkeys compared with rodents; the explanation for this dif-
`ference is unknown. After multiple doses, a decrease in clear-
`ance resulted in an increase in the terminal elimination half-life
`(t1/2) and AUC (3– 4-fold) in rats and cynomolgus monkeys,
`suggesting drug accumulation. Even though there were no gen-
`der differences in systemic exposure, it appears that female
`decedent rats exhibited a greater degree of toxicity compared
`with males based on the number and types of lesions, as well as
`on the total number of unscheduled deaths. Using an ex vivo 20S
`
`proteasome assay to measure inhibition of the chymotrypsin-
`like proteolytic activity in whole blood cells, proteasome inhi-
`bition increased with dose and returned to baseline by about
`72 h in rats and monkeys. After a single dose of [14C]bort-
`ezomib, bortezomib-related radioactivity was eliminated slowly
`from tissues (with highest concentrations in liver and kidneys);
`incomplete recovery of administered radioactivity in rats and
`monkeys suggests extensive tissue distribution and retention of
`bortezomib and its metabolites. Radioactivity was detected in
`the brain of monkeys but not of rats.
`Cardiovascular safety pharmacology studies conducted in
`cynomolgus monkeys showed that the administration of dosages
`ⱖ3.0 mg/m2 resulted initially in physiologically significant
`heart rate elevations, then profound progressive hypotension,
`bradycardia, and death 12–14 h postdose. Additional studies in
`monkeys showed bortezomib increased heart rate (ⱖ1.2 mg/
`m2), decreased mean arterial pressure (ⱖ2.4 mg/m2), increased
`ventricular contractility (ⱖ3.6 mg/m2), and increased cardiac
`output (ⱖ3.6 mg/m2). Mortality was not reported in this study;
`however, this study is inadequate to address drug-associated
`mortality observed in the previous studies because these mon-
`keys were sacrificed before signs of terminal hypotension and
`imminent mortality occurred. Bortezomib-related radioactivity
`was distributed to the myocardium. Histopathological findings
`in repeat-dose monkey studies showed cardiac necrosis at doses
`ⱖ0.9 mg/m2. Whether the observed cardiac effects are depend-
`ent on local drug disposition and/or direct drug-myocardial
`toxicity is unknown.
`Bortezomib exhibited clastogenic activity in the in vitro
`chromosomal aberration assay using Chinese hamster ovary
`cells but was not genotoxic when tested in the in vitro mutage-
`nicity assay (Ames test) or the in vivo micronucleus assay.
`Teratological effects were examined in the rat and the rabbit. No
`formal evaluation of fertility or peri- and postnatal development
`(Segments I and III, respectively) were conducted. Pregnant
`rabbits given bortezomib during organogenesis at a dose of 0.6
`mg/m2 experienced significant postimplantation loss and a de-
`creased number of live fetuses at minimally maternally toxic
`doses. Live fetuses also showed significant decreases in fetal
`weight. This dose is approximately one-half the clinical dose
`(1.3 mg/m2). On the basis of embryo lethality findings in rats
`and rabbits, and the effects on primary and secondary sex organs
`observed in the 6-month rat study and the 9-month monkey
`toxicity studies, bortezomib is likely to have an adverse effect
`on pregnancy. However, bortezomib was not teratogenic in rats
`and rabbits at the highest dose tested, 0.5 mg/m2 in the rat and
`0.6 mg/m2 in the rabbit, when administered during organogen-
`esis. These dosages also are approximately one-half of the
`human clinical dose. Bortezomib is labeled “Pregnancy category
`D;” because of the potential of significant adverse effects on the
`developing fetus, women are strongly advised not to become
`pregnant while taking bortezomib.
`
`CLINICAL STUDIES SUPPORTING APPROVAL
`Three Phase I dose finding trials of bortezomib as mono-
`therapy were performed in a total of 123 patients with a variety
`of advanced malignancies. Two Phase II studies were performed
`in 256 patients with multiple myeloma who had not achieved a
`
`Downloaded from on September 23, 2015. © 2004 American Association forclincancerres.aacrjournals.org
`
`
`
`Cancer Research.
`
`ANACOR EX. 2012 - 4/12
`
`

`
`3958 Bortezomib in Multiple Myeloma
`
`response to, or who had relapsed after, initial therapy. If patients
`progressed after two cycles or experienced no improvement
`after four cycles, dexamethasone 20 mg daily p.o. for 2 days was
`added to each bortezomib dose. An extension studies allowed
`continued therapy in those patients who appeared to benefit. The
`extension study provided safety information on longer-term
`therapy and efficacy information on response duration.
`
`PHASE I STUDIES
`The Phase I studies were performed in patients with ad-
`vanced malignancies using weekly and twice-weekly dosing
`schedules. Weekly dosing was associated with dose-limiting
`toxicities of diarrhea, hypotension, tachycardia, and syncope
`with doses ⬎1.6 mg/m2. The maximum tolerated dose was
`found to be 1.3 mg/m2 when given twice-weekly in the first 2
`weeks of a 3-week cycle. This compares with a maximum
`tolerated dose of 1.04 mg/m2 when given twice-weekly for the
`first 4 weeks in a 6-week cycle. At the 1.3 mg/m2 dose, the 1-h
`mean percentage proteasome inhibition measured in patients’
`WBCs on day 1, cycle 1, is higher than the corresponding value
`at cycle 7 (70.5 versus 55%). The relationship between protea-
`some inhibition and dose suggests that the optimal bortezomib
`dose may be between 1.0 mg/m2 and 1.3 mg/m2. The twice-
`weekly dosing each 21-day cycle was selected because ⬃25%
`more drug could be tolerated.
`
`PHASE II CLINICAL STUDIES
`Two Phase II studies assessed the safety and efficacy of
`bortezomib. A small, open-label, randomized Phase II dose-
`finding study, Clinical Response and Efficacy Study of bort-
`ezomib in the Treatment of
`relapsing multiple myeloma
`(CREST) was performed in 54 patients with relapsed multiple
`myeloma to provide some dose-response data (46). The 1.3-
`mg/m2 dose was compared with the 1.0-mg/m2 dose using a
`21-day cycle with treatment given during the first 2 weeks. The
`sponsor chose the higher dose because of a somewhat higher
`overall response rate that included minimal responders. The
`larger Study of Uncontrolled Multiple Myeloma managed with
`proteasome Inhibition Therapy (SUMMIT) was an open-label,
`single-arm, multicenter study of patients who had received at
`least two prior therapies and demonstrated disease progression
`on their most recent therapy (15). Patients were eligible if they
`had relapsed after a response to standard first-line chemotherapy
`(e.g., vincristine-doxorubicin-dexamethasone or melphalan-
`prednisone) or high-dose chemotherapy, and were refractory
`(i.e., failure to achieve at least CR, PR, or stable disease) to their
`most recent chemotherapy. Primary refractory patients were not
`enrolled.
`Bortezomib 1.3 mg/m2 was administered by i.v. bolus over
`3–5 s on days 1, 4, 8, and 11 in a 21-day cycle. A maximum of
`eight cycles (24 weeks) was planned, but treatment could be
`continued for responding patients in a continuation study. Treat-
`ment was withheld in patients experiencing ⱖ grade 3 nonhe-
`matological or grade 4 hematological toxicities until resolution
`or grade 1 was attained, and treatment was then resumed at the
`next lower dose level, either 1.0 or 0.7 mg/m2. Patients with
`progressive disease after completing two cycles or who experi-
`enced no response after four cycles could be treated with the
`
`addition of dexamethasone, 20 mg p.o. (each day of, and the day
`after, bortezomib administration, i.e., 40 mg with each dose).
`These patients were analyzed separately for efficacy and were
`not included in the primary analysis. Safety assessments per-
`formed during treatment
`included monitoring for adverse
`events, a directed questionnaire for neurological toxicities, spe-
`cialized neurological testing, and clinical examinations.
`The SUMMIT trial enrolled 202 patients. Eighty-four %
`had IgG or IgA myeloma and advanced disease at diagnosis, and
`80% had a Karnofsky performance-status score ⱖ80. The mean
`age was 59 years; 81% were white, 10% were black, and 60%
`were male. Ninety-two % had been treated with three or more of
`the major classes of agents used to treat myeloma (steroids,
`alkylating agent, thalidomide, or anthracyclines). The median
`number of previous therapies was six (range, 2–15). Sixty-four
`% had received high-dose therapy and stem-cell transplantation.
`Five patients had not been treated with cytotoxic chemotherapy;
`these were excluded from the efficacy analysis. In comparison,
`the CREST study enrolled a less heavily pretreated population;
`the mean number of prior therapies was three, compared with
`six in the larger study. The mean Karnofsky performance-status
`score was also higher in the CREST study; otherwise, the trial
`characteristics were similar. Baseline patient and disease char-
`acteristics for both studies are summarized in Table 2.
`
`RESULTS
`The primary objective of the Phase II studies was the
`determination of overall response rate (CR ⫹ PR ⫹ minimal
`response). Responses were assigned by an Independent Review
`Committee based on the EBMT criteria described above (sec-
`tion “Response Criteria in Multiple Myeloma”). PRs required a
`50% reduction in serum M protein and 90% reduction in urine
`M protein. Additional response analyses including remission by
`SWOG criteria and rate of complete resolution of M protein by
`PEP were performed to facilitate comparison with other reports
`of available therapy (see Table 1.) Minimal responses were not
`included in the United States Food and Drug Administration
`(FDA) analysis because these responses were deemed less likely
`to predict clinical benefit. All of the responses required confir-
`mation at 6 weeks by protein electrophoresis (CRs required
`repeat IF also.)
`Fifty-four patients were enrolled in the CREST study. The
`response rate (CR⫹PR) was 38% in the 1.3-mg/m2 group com-
`pared with 30% in the 1.0-mg/m2 group. One patient in each
`group experienced a CR by EBMT criteria, and two additional
`patients in the 1.0-mg/m2 group experienced complete resolu-
`tion of myeloma M protein by PEP. If minimal responses were
`included, the (CR ⫹ PR ⫹ minimal response) rate was 50% in
`the 1.3-mg/m2 group compared with 33% in the 1.0-mg/m2
`group. This observation of a higher response rate led the sponsor
`to recommend the higher dose for further study, although the
`numbers were too small for statistical dose-response compari-
`sons.
`In the SUMMIT study, the FDA analysis identified 188
`patients of the 202 enrolled who had evaluable disease and who
`fulfilled all eligibility criteria. The study population included
`patients with numerous adverse prognostic features including ␤
`2
`microglobulin levels above 4 mg/liter, cytogenetic abnormali-
`
`Downloaded from on September 23, 2015. © 2004 American Association forclincancerres.aacrjournals.org
`
`
`
`Cancer Research.
`
`ANACOR EX. 2012 - 5/12
`
`

`
`Table 2 Summary of characteristics of patients enrolled in the
`bortezomib Phase II studies in multiple myeloma
`
`CREST dose level
`
`1.0 mg/m2
`(n ⫽ 28)
`
`1.3 mg/m2
`(n ⫽ 26)
`
`SUMMITa
`dose level
`1.3 mg/m2
`(n ⫽ 202)
`
`Characteristic
`
`14 (50)
`14 (50)
`
`25 (89)
`3 (11)
`0
`0
`
`65
`39, 82
`
`2 (7)
`1 (4)
`8 (29)
`17 (61)
`
`9 (35)
`17 (65)
`
`20 (77)
`3 (12)
`2 (8)
`1 (4)
`
`61
`30, 84
`
`2 (8)
`2 (8)
`8 (31)
`14 (54)
`
`2
`2
`⬍0.5–20 ⬍0.5–8
`7/24
`11/23
`2/7
`3/11
`14/24
`11/23
`
`121 (60)
`81 (40)
`
`164 (81)
`21 (10)
`5 (2)
`12 (6)
`
`59
`34, 84
`
`19 (10)
`21 (11)
`74 (38)
`82 (42)
`
`4
`1–18
`60/172
`26/172
`138
`
`Clinical Cancer Research
`
`3959
`
`CRs or PRs to bortezomib monotherapy (see Table 3.) Among
`the 47 PRs, there were 12 patients who exhibited complete
`resolution of M protein by PEP but not by IF.
`In addition to the protocol-specified response analysis
`(EBMT), a separate analysis by SWOG criteria showed clinical
`remission criteria were fulfilled by 33 patients

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