throbber
Novel Therapeutic Agents for the Treatment
`of Myelodysplastic Syndromes
`
`Bruce D. Cheson, James A. Zwiebel, Janet Dancey, and Anthony Murgo
`
`activity
`agents have demonstrated
`Few chemotherapy
`in patients with myelodysplastic syndromes (MOS) and
`supportive management
`remains
`the standard
`of care.
`An increasing number of new drugs in development are
`being directed
`at specific molecular
`or biological
`tar-
`gets of these diseases. Topctecan, a topoisomerase I
`inhibitor,
`has shown single-agent
`activity and is now
`being
`combined
`with
`other
`agents,
`including
`cy~
`tarablne. The aminothiol
`amifostine
`induces
`responses
`in about 30% of patients; however,
`its role is still being
`clarified. Agents
`that
`inhibit histone
`deacetylase
`and
`target DNA hypermethylation,
`thus permitting
`dere-
`pression of normal genes,
`include 5-azacytidine,
`decita-
`bine, phenyl butyrate.
`and depsipeptide. Arsenic triox-
`ide has demonstrated
`impressive
`activity
`in acute
`promyelocytic
`leukemia
`and preclinical
`data
`suggest
`the potential
`for activity in MOS. UCN~OI
`is a novel
`agent
`that
`inhibits protein kinase C and other protein
`kinases important
`for progression
`through
`the G. and
`G2 phases of the cell cycle. Dolastatfn-In has extremely
`potent
`in vitro activity against a variety of tumor
`cell
`lines. Since its dose-limiting toxicities
`include myelosup-
`pression,
`it
`is being studied
`in acute myelogenous
`leukemia (AML) and MOS. Rat may playa role in MOS.
`and activation
`of this gene and its signaling pathways
`may require farnesylation.
`Several
`farnesyl
`transferase
`inhibitors
`are now available for study in patients with
`MOS. An increasing
`body of data suggests
`a possible
`role for angiogenesis
`in MOS. and several antlanglcgen-
`esis agents are in clinical
`trials,
`including thalidomide.
`SU5-416. and anti-vascular
`endothelial
`growth
`factor
`(VEGF) antibodies. Development
`of new drugs
`and
`regimens will be facilitated by recently developed stan-
`dardized response
`criteria. Future clinical
`trials should
`focus on rational
`combinations
`of these
`agents
`and
`others with the goal of curing patients with MOS.
`Semin Oneal 27:560-577. This is a US government work.
`There are no restrictions on its use.
`
`THE MYELOOYSPLASTlC syndromes (MOS)
`
`are a heterogeneous group of hematopoietic
`disorders characterized by pancytopenia,
`generally
`in the setting of a hypercellular bone marrow. MDS
`
`From the Cancer Therapy Evaluation Progrcm (CTEP), Divi~
`sion of Cancer Treatment and Diagnosis, National Cancer
`Institute,
`Bethesda, MD.
`to Bruce D. Cheson, MD, National
`requests
`Address
`reprint
`Cancer Institute, Executive Plaza North-Room 741, Bethesda, MD
`20892.
`This is a US government work. There are no restrictions on its
`""
`
`have historically been referred to as oligo blastic
`leukemia, refractory anemia, smoldering acute leu-
`kemia, or preleukemia.
`In 1982,
`the French-
`American-British
`(FAB) group presented a classifi-
`cation, modified in 1985, which currently is the
`most widely used.t- The FAB group separated
`MOS into five categories: refractory anemia (RA),
`RA with ringed sideroblasts
`(RARS), RA with
`excess blasts (RAEB), and RAEB in transforma-
`tion (RAEB- T). The distinction between RAEB- T
`and acute myelogenous
`leukemia (AML)
`is based
`on histopathology, not clinical features, As a result,
`patients with MDS may exhibit a clinical picture
`consistent with AML with rapidly increasing num-
`bers of blasts. but without
`the requisite number
`to
`fulfill
`the
`criteria
`for
`the diagnosis of AMLJ
`Recently, a World Health Organization
`(WHO)
`steering committee proposed changes to the MDS
`subtypes with the major modifications
`including
`reclassifying chronic myelomonocytic
`leukemia
`(CMML)
`as a myeloproliferative
`disorder
`and
`decreasing the threshold for diagnosing AML from
`30% blasts to 20%4 This system may eventually
`replace the FAB.
`The likelihood of transformation to AML varies by
`FAB subtypes-s:approximately 10% to 20% for RA or
`RARS, 20% to 30% for CMML, 40% to 50% for
`RAEB, and 60% to 75% for RAEB-T. Nevertheless,
`the MDS are uniformly fatal, even without progression
`to AML, because of infection and bleeding.'·!O
`Over the years, a number of scoring and prognos-
`tic systems have been published to facilitate com-
`parisons among reports of various treatments
`for
`MOS_ Recently,
`the International Prognostic Scor-
`ing System (lPSS) has been widely adopted.'!
`Factors
`taken into consideration
`included bone
`marrow blasts, cytogenetics, and cvropenias. Groups
`were identified with relative risks for transforma-
`tion to AML and overall survival. Patients
`in the
`good cytogenetics group were those with a normal
`karyotype; poor risk included patients with com-
`plex abnormalities
`or with an involved chromo-
`some 7; intermediate-risk
`patients consisted of all
`others (Table 1)-
`There are no curative therapies other than stem
`cell transplantation, which is an option for only a
`subset of patients. Therefore, numerous
`therapies
`
`560
`
`Seminars in Oncology, Vol 27, No 5 (October),
`
`2000: pp 560·577
`
`DR. REDDY’S LABS., INC. EX. 1029 PAGE 1
`
`

`

`NEW THERAPEUTIC
`
`AGENTS FOR. MDS
`
`561
`
`Table
`
`I.
`
`International
`
`Prognostic
`
`Scoring
`
`System for
`
`the Myelodysplastic
`
`Syndromes
`
`Prognostic Variable
`
`Bone marrow blasts (%)
`Karyotype
`Cyropanlas
`
`(no. of lineages)
`
`0
`
`<5
`Good
`0/1
`
`0.5
`
`5-10
`Intermediate
`2/3
`
`Score
`
`1.0
`
`Poor
`
`1.5
`
`11-20
`
`2.0
`
`21·30
`
`NOTE. The total of the values for each prognostic
`variable is used to place patients
`high. These risk groups have significantly different outcomes.
`
`in , of 4 risk groups:
`
`low, intermediate-]
`
`, ineermedraee-z.
`
`and
`
`to improve
`have been and are being investigated
`the outlook for these patients. Drugs selected for
`study in MOS have
`typically been those with
`significant activity in AML Thus, cytarabine has
`been most widely evaluated,
`dating
`back more
`than 30 years when Ellison et a[1' first reported
`complete
`remissions with doses of cytarabine
`as
`low as 10 mg/m'/d. Response
`rates were clearly
`dose-dependent, which encouraged
`the develop-
`ment of higher dose regimens. Subsequently, anec-
`dotal
`reports and small series were published in
`which cytarabine
`at 10% to 20% of the standard
`dose administered
`either
`subcutaneously
`or by
`continuous
`intravenous
`infusion appeared to be
`effective in the treatment of AML and MOS."·"
`Additional
`studies and a randomized phase III trial
`failed to support a major role for this therapy.z3.z5
`Anthracyclines
`and related compounds
`have
`had been srudied as single agents only to a limited
`In a study in which hydroxyurea
`and
`extent.26,Z7
`etoposide were compared in patients with CMML,
`response rates and survival were not
`impressive
`with either agent, but favored the former.28 Other
`drugs that have been evaluated include S-tluogua-
`nine and homoharringtonine,
`but both showed
`limited activity.29,JO
`
`NEW AGENTS
`Several agents with unique mechanisms of activ-
`ity are currently
`or will soon be evaluated
`in
`clinical
`trials for patients with MOS.
`Topoisomerase IInhibitors
`Topotecan is a topoisornerase I inhibitor whose
`activity in acute
`leukemia
`led to its testing in
`MOS. The initial report
`included 47 patients with
`RAEB, RAEB- T, or CMML.JI They were a poor-
`risk group, as demonstrated
`by the fact
`that
`the
`median age was 66 years, 70% exhibited cytoge-
`netic
`abnormalities,
`and more
`than
`half were
`
`before topotecan therapy. Topo-
`thrombocytopenic
`tecan was delivered at a dose of 2 mg/m2 as a
`continuous 24~hour infusion for 5 days. Treatment
`resulted in 28% complete remissions and an addi-
`tional13% of patients who experienced significant
`hematologic
`improvement. All eight patients with
`cytogenetic
`abnormalities
`before treatment
`and
`who achieved a complete remission became cvtoge-
`netically normal once in complete remission. The
`median remission duration was 7.5 months with
`38% of patients
`still alive 1 year following treat-
`ment. Whether
`chronic oral topotecan is effective
`is undergoing evaluation.
`The same investigators have shown that cornbi-
`nation of topotecan
`and cvtarabine
`is extremely
`active in patients with MDS. Beran et aP2 reported
`on 86 patients with MOS and CMML, most of
`whom (66%) were previously untreated, but who
`were considered high risk based on age or cvroge-
`netic abnormalities. Topotecan was administered
`ar a dose of 1.25 mg/m' by continuous
`infusion
`daily for 5 days, and cytarabine
`at 1 glm' by a
`2-hour
`infusion
`daily for 5 days. A complete
`remission was attained in 56% of patients, with 7%
`treatment-related
`deaths and a median survival of
`60 weeks."
`Preliminary
`results have been pub-
`lished of aggressive combination
`of
`topotecan,
`fludarabine,
`cytarabine,
`and granulocyte
`colony-
`stimulating factor (G-CSF);
`there were 50% com-
`plete remissions and 40% partial
`remissions, and
`the regimen appeared to be well rolcrarcd.P
`
`Amifostine
`(Ethyol; Alza Pharmaceuricals, Palo
`Amifostine
`is a phosphorylated
`aminothiol
`that
`Alto, CAl
`protects bone marrow progenitors
`and other nor-
`mal
`tissues
`from the
`toxicities
`associated with
`chemotherapy
`or radiation therapy.
`It was devel-
`oped by the Walter Reed Army Medical
`Institute
`
`DR. REDDY’S LABS., INC. EX. 1029 PAGE 2
`
`

`

`S62
`
`during
`the military code name WR-2721)
`(thus,
`the Cold Wat as part of a classified research project
`to identify an agent that would protect military
`personnel
`from radiation in the event of nuclear
`war. Arnifostine was found to afford greater protec-
`tion against radiation than more than 4,000 other
`compounds
`screened. Nevertheless,
`the Army ter-
`minated development
`of this compound in 1988
`because of its poor oral bioavailability
`and the
`prohibitive nausea, vomiting, diarrhea, and abdomi-
`nal cramps with the oral formulation.
`Further research was encouraged by the observa-
`tion that amifostine stimulates hematopoiesis in
`both animal models and in vitro studies, and that
`it
`enhances
`the formation of hematopoietic
`progeni-
`tors from MDS bone marrow. In the initial phase
`1/11studv," the drug was administered at doses of
`100,200, or 400 mg/m' three times per week or 740
`investigators
`mg/m2 weekly for 3 weeks. These
`treated 18 patients at a median age of 73 years.
`FAB types included RA (seven patients), RARS
`(n = 5), RAEB (n = 4), and RAEB-T (n = 2).
`Seventeen patients were anemic, 15 of whom were
`transfusion-dependent,
`12 had an absolute neutro-
`phil count less than 1,000{pL and 14 were thrombo-
`cytopenic. Hematologic
`improvement was ob-
`served
`in 83% with
`the
`three-times-a-week
`schedules,
`including either an increase in neutro-
`phils or a reduction in red blood cell transfusion
`requirements. More than 40% of patients had a rise
`in their platelet counts. However,
`there was accel-
`eration to AML in several patients with RAEB~T.
`Although 61% of patients had clonal cyrogenetic
`abnormalities
`before therapy,
`the
`abnormalities
`persisted even in patients with a hematologic
`response. No data regarding duration of response
`were provided, although responses were reported to
`persist during continuation therapy.
`List et ap5 reported the results of a subsequent
`multicenter
`trial of arnlfostine in 117 patients, 104
`of whom were evaluable at the time of presenta-
`tion. A neutrophil
`response occurred in 10 (33%)
`of 30 patients, and was considered major
`in nine
`and minor in the other. A red blood cell response
`was evaluable in 66 patients, and a major response
`occurred in seven, with three experiencing a minor
`response. A major improvement
`in platelet count
`was seen in seven of 27 patients, with a minor
`response in three others, and 21% of patients had
`an increase in the reticulocyte count. A decrease in
`myeloblasts and sideroblasts occurred in 28% and
`
`CHESON ET AL
`
`response rate was
`respectively. The overall
`31%,
`30%, which is significantly
`lower
`than
`in the
`previous trial. Adverse events that were moderate
`or severe included fatigue
`(14%, 18%), nausea
`09%,
`36%),
`and vomiting
`04%,
`27%).
`In a
`smaller series."
`a single or multilineage
`response
`was noted
`in five of 12 patients
`(58%). The
`absolute neutrophil
`count
`Increased in 25% (by
`102 to 1,560{pL), platelets
`in 50% (by 24,000 to
`49,000{pL), reticulocytes
`in 25% (1.9% to 20%),
`and hemoglobin in 16% (5.3 to 5.6 g{dL).
`In other
`reports,
`results with this agent were
`disappointing.37,38 Hofmann
`et ap8 described 32
`patients with RA/RARS
`(n = 26) and RAEB{
`RAEB-T (n ~ 15) treated at a dose of 200 mg{m'
`three times per week followed by a Zvweek interval,
`for four courses. Limited benefit was observed even
`in patients with low- or intermediate-risk
`disease
`by the IPSS.
`in MDS is still being
`The role of amifostine
`clarified. Nevertheless,
`combinations of amifosrine
`with other agents such as 'i-azacytidine
`are being
`evaluated.
`
`Agents That Target Transcription
`the rno-
`Recent developments
`in understanding
`lecular basis
`for
`transcriptional
`repression
`and
`activation
`have presented
`new possibilities
`for
`cancer therapy. Two mechanisms of gene silencing,
`promoter hvpermethvlauon
`and histone deacetvla-
`tion,
`appear
`to be interrelated.
`The
`utility of
`targeting DNA hypermethylation
`and histone
`deacetylation
`is being explored clinically. Agents
`shown to inhibit
`histone
`deacetylase
`in vitro
`include sodium phenylbutyrate,
`depsipeptide, hy-
`brid polar compounds.'?
`and MS-27-275.40 Hypo-
`methylating agents include 5~azacytidine and 'i-aza-
`Zcdeoxycyudine. The exploration
`of these agents
`in the clinic, either alone or in combination with
`retinoids, demethylation
`agents, and chemothera-
`is a novel and promising area of
`peutic agents I
`cancer therapeutics.
`agents.
`Hypomethylating
`and
`5-Azacytidine
`fi-aaa-Z." deoxycytidine are pyrimidine analogs that
`have been extensively evaluated in patients with
`MDS. These compounds are metabolized intracel-
`lularly to triphosphates
`and subsequently incorpo-
`rated into newly synthesized DNA, where they
`directly inhibit DNA synthesis and inhibit
`the
`activity of DNA rnethvlrransferase,
`the enzyme
`required for 5/ -cytosine methylation
`of cytosine-
`
`DR. REDDY’S LABS., INC. EX. 1029 PAGE 3
`
`

`

`NEW THERAPEUTIC AGENTS FOR MDS
`
`56l
`
`As a result,
`(CpG) dmuclcondes.tl"
`guanosine
`cytosine methylation
`is blocked in newly repli-
`cated DNA, but not
`in the DNA of resting at
`nondividing
`cells.
`Inhibition
`of methylation
`by
`5~azacytidille and decitabine
`is associated with
`transcription of genes previously silenced by meth-
`ylation of promoter region CpG;rich islands, and
`with cellular phenotypic changes; these effects can
`occur at concentrations
`that are too low to inhibit
`DNA synthesis directly or to cause substantial
`cytotoxicity.41,43-45The potential application of
`'i-azacyrtdine and decitabine as inhibitors of DNA
`methylation and inducers of cell differentiation of
`normal and neoplastic hematopoietic progenitor
`cells is an area of active investigation.43,45-48
`5~Azacytidine initially demonstrated activity in
`AML,49-53but with considerable toxicity at doses
`required for response. Since the drug also induces
`in vitro cellular differentiation in association with
`hypomethvlation
`of DNA,
`it was of interest
`for
`study in MDS. Chitambar
`et a)54used a relatively
`low dose (10 to 35 mg/m'/d for 14 days) to treat 13
`patients,
`three of whom achieved a partial rei
`sponse. Cancer and Leukemia Group B (CALGB)
`conducted a phase II trial of S-aaa-
`mvestigarors'?
`cytidine at 75 mgfm'/d by continuous
`infusion for 7
`days every 28 days in 48 patients with MDS and
`noted 11% complete remissions and 25% partial
`remissions. Major toxicities
`included nausea and
`vomiting: one patient died of neutropenic sepsis.
`Subcutaneous administration resulted in slightly
`lower response, rates-7% complete
`remissions,
`17% partial remissions, and 14% with trilineage
`improvement, but less than a partial response.S
`These findings are similar to those achieved with
`low-dose cytarabine.
`The CALGB recently reported the pteliminary
`results of a phase III randomized trial of 5;azacyti;
`dine versusobservation in 191 patients with MDS.57
`The patients were stratified by FAB subtype (19%
`RA, 4% RARS, 42% RAEB, 21% RAEB-T, 6%
`CMML);
`patients with RA or RARS had,
`in
`addition, symptomatic cvropenias.
`'i-Azacyridine
`was administered subcutaneously at a dose of 75
`mg/m'/d fat 7 days every 4 weeks for four cycles.
`Patients on the observation arm could receive
`'i-azacytidine upon progression. Hematologic
`rei
`sponses were significantly higher in patients ran;
`domized to receive 5;azacytidine compared with
`(P < .0001): 63% (6% complete
`observation
`re-
`sponse, 10% partial response, and 47% improve-
`
`merit) versus 7% (all improvement, no complete or
`partial responses). The median time to leukemic
`transformation or death was 22 months for patients
`on the treatment arm, compared with 12 months
`for
`the patients
`randomized
`to observation
`(P = .0034). The 12- and 24-month
`overall sur-
`vival rate was higher in patients randomized to
`receive azacytidine (70% and 4 I % versus 62% and
`25%, respectively), as was the median survival
`time (I8 versus 14 months),
`but
`the differences
`were not yet significant. Treatment with 5~azacyti-
`dine was associated with subjective improvement
`in quality of life as measured by fatigue, dyspnea,
`physical functioning, positive affect, and psycho-
`logic distress.V Whether 5;azacytidine
`improves
`overall survival or reduces transformation to leuke-
`mia will require additional follow-up evaluation.
`5-Aza-2' deoxycytidine
`(decitabine)
`is another
`hypomethylating agent with potent in vitro activ-
`icy.In earlier studies, decitabine administered as an
`intermittent
`intravenous infusion achieved brief
`responses in a small series of patients with MDSj
`however, the majority experienced life-threatening
`neutropenia
`and/or thrornbocvroperua.t? Wijer;
`mans et al60 reviewed the experience with this
`agent
`in MDS and found a 54% tesponse rate of29
`elderly patients,
`although
`there were 17% toxic
`deaths. This drug is under developmenr
`for MDS
`both in Europe and the United States.",60
`Histone deacetylation and DNA hypermethylation.
`Retmoids, other hormone receptors, and the Myel
`Mad/Max network of growth regulators exert their
`effects on gene expression by interacting with
`nuclear corepressor complexes that are present on
`the DNA of promoter regions.61,61 Gene silencing
`occurs with the recruitment ofhistone deacetylases
`and the formation of a nuclear corepressor-histone
`deacetylase complex (NCHDC). Histone deacetv-
`lase catalyzes the removal of acetyl groups from
`histone proteins,
`inducing a conformation change
`that results in an environment unfavorable to gene
`transcription. A NCHDC has been found to play
`an important role in acute promyelocytic leukemia
`(APL), where the NCHDC is recruited by both the
`PML-RARa
`and PLZF-RARa
`fusion
`proteins,
`which form as a consequence
`of chromosomal
`trans locations
`t(l5; 17)
`and
`t(l I; I 7),
`respec-
`tively6J.67 A NCHDC is also recruited by ETO, a
`component of the fusion product resulting from
`the t(8;21) chromosomal
`translocation in AML.68,69
`Moreover,
`inhibitors of histone deacetylase have
`
`DR. REDDY’S LABS., INC. EX. 1029 PAGE 4
`
`

`

`564
`
`CHESON ET AL
`
`repression
`been found to overcome transcriptional
`and to potentiate
`retinoid-induced
`differentiation
`of APL and AML cells.63•66•69 A clinical
`test of this
`observation was performed in a patient with APL
`who had become tefractory to both chemotherapy
`and all-tram retinoic
`acid (ATRA). Administra-
`rion of both ATRA and a histone
`deacetylase
`inhibitor,
`sodium phenylbutyrate
`(see below),
`re-
`sulted in a complete remission. The clinical re-
`sponse was associated with acetylation of histone
`proteins in the leukemic cells."?
`in gene
`While methylation
`of CpG islands
`promoter
`regions has
`long been known
`to be
`associated with gene silencing,
`it was not known
`how such DNA hvpermerhvlauon
`exerts its effect
`on gene transcription. Recent studies have shed
`light on both the role of DNA hypermethylation
`in the inactivation
`of tumor suppressor genes, as
`well as the mechanism of transcriptional
`repres-
`sion. Examples of genes associated with CpG
`hypermerhvlation
`include, among others, RB in
`retinoblastoma, VHL (the von Hippel-Lindau gene)
`p16lNK4A
`and p15lNK4A
`in renal carcinoma,
`(cvclin-dependenr
`kinase inhibitors)
`in solid tu-
`mors and in hematologic malignancies, and hMLHl
`(a DNA mismatch repair gene) in colon cancer."
`The mechanism of gene silencing by DNA hyper-
`methylation
`now appears to involve the recruit-
`ment of a NCHDC by the methyl-Cpff-hinding
`protein, MeCP2.72,73 In fact, the combined admin-
`istration of a demethylaring
`agent and a histone
`deacetvlase inhibitor has been shown to svnergiae
`in reactivating genes that were silenced in cancer
`cells.74 This finding nor only links the processes of
`DNA hypermethylation
`and histone
`deaceryla-
`tion, but
`also presents
`therapeutic
`targets
`for
`agents
`that
`are relatively nontoxic,
`or used at
`nontoxic doses.
`
`Fhenylbutyrate
`(FB) is a low-molecular-weight
`Phenylbutyrate
`phenyl-farty acid that been used clinically to treat
`hyperammonemia
`in children with inborn errors of
`urea synthesis.75 It also been shown to enhance
`fetal hemoglobin production in some patients with
`hemoglobmopathies.ts A number of mechanisms
`have been proposed for the antitumor effect of PB,
`including (1) elimination
`of glutamine necessary
`for nucleic acid and protein synthesis in rapidly
`growing normal and tumor cells77,78;(2) inhibition
`of the mevalonate pathway of cholesterol synthesis
`
`process-
`leading to interference of post-translational
`ing of proteins, modification of lipid metabolism,
`inhibition
`of protein isoprenylation, and regula-
`tion of gene expression through DNA hvpomethyl-
`ation 79,80;(3) activation of a peroxisome prolifera-
`tor-activated
`receptor
`by PB, a transcriptional
`factor regulating lipid metabolism and cell growrh'":
`and (4)
`regulation
`of gene expression
`through
`histone hyperacetylation
`via inhibition of nuclear
`histone deacetylases.82-84
`PB has been shown to induce differentiation,
`tumor cytostasis. and reversion of malignant pheno-
`type in several
`in vitro models.80,85-89 PB, as a
`histone deacetylase inhibitor, may have synergistic
`activity with ATRA in the treatment of APL.65,67,90
`The PML-RAR fusion protein was shown to re-
`cruit a transcriptional
`corepressor
`complex that
`includes a histone deacetvlase. ATRA alone could
`partially
`dissociate
`the
`complex,
`allowing
`in-
`creased transcription, but butyrate (or other inhibi-
`tors of histone deacetylases)
`in combination with
`ATRA was able to completely abrogate the inhibi-
`tion of transcription.
`In light of these observations,
`an APL patient who experienced multiple relapses
`after ATRA treatment was treated with PB in
`combination with ATRA under
`compassionate
`release, and achieved a complete remission.i''
`
`Depsipeptide
`is a bicyclic pep-
`(NSC 630176)
`Depsipepride
`tide originally isolated from Chromobacterium viola~
`cetcn, strain 968, by Fujisawa Pharmaceutical Co
`(Osaka, Japan). In the original observations, depsi-
`peptide selectively decreased the mRNA expres-
`sion of the c-myc oncogene
`and inhibited
`the
`growth of the Ha-ras-transfortned NIH3T3 clonal
`cell
`line, Ras-I. but had no effect on Ha-res
`mRNA expression.vt It did not affect DNA synthe-
`sis, but caused cell cycle arrest at GO/Gl. Recently,
`it has been shown to be a histone
`deacerylase
`mhibiror"
`of
`incubation
`that
`Byrd et al demonstrated
`chronic lymphocytic leukemia cells with depsipep-
`tide resulted in an alteration
`in apoptosis-assoct-
`ated proteins: an increase in Bax with no change in
`Bcl-Z, and a decrease in p27 expression."
`In collaboration with Fujisawa Pharmaceutical
`Co,
`the National Cancer
`Institute
`(NCI)
`is cur-
`rently sponsoring two phase I trials of depsipeptide
`administered as a -l-hour intravenous
`infusion.
`In
`one trial, a once-weekly infusion schedule (days 1,
`
`DR. REDDY’S LABS., INC. EX. 1029 PAGE 5
`
`

`

`NEW THERAPEUTIC AGENTS FOR MDS
`
`565
`
`8, and 15 every 28 days) is used, while the other
`trial evaluates a twice-weekly (days 1 and 5 every
`21 days) schedule.
`
`MS-27-275
`that was
`MS-27-275 is a benzamide derivative
`(Tokyo,
`synthesized
`by Mitsui Pharmaceuticals
`Japan) in a search for novel antitumor agenrs.w
`The compound was found to have histone deacery-
`lase activity in vitro at micromolar concentrations.
`In addition, when administered orally, MS-27-275
`inhibited the growth of a number of tumor xeno-
`grafts. The NCI,
`in collaboration with Mitsui
`Pharmaceuticals, plans to sponsor phase I trials of
`this agent in the near future.
`
`Hylrrid Polar Compounds
`(HMBA) was the
`Hexamethylene
`bisacetamide
`first of the class of hybrid polar compounds
`to be
`evaluated as an antitumor agent
`in MDS and
`AML. The limited clinical activity that was ob-
`served was attributed to the inability to achieve
`the plasma concentrations
`that were required to
`induce differentiation in cells
`in vitro and to
`Subsequently,
`dose-limiting
`rhrombocytopenta.P'
`Richon et aP9 described compounds structurally
`related
`to HMBA,
`but which
`exhibited
`3-log
`greater potency in inducing terminal differentia-
`In
`tion and apoptosis in transformed cell
`lines.
`addition, these compounds possess histone deacety-
`lase inhibitory activity at micromolar concentra-
`tions. Recently, one such compound, Mvcarboxv-
`cinnamic acid bishydroxamide (CBHA), was found
`to induce apoptosis in human neuroblastoma, and
`the effect was associated with CD95jCD95
`ligand
`expression by the tumor cells." These agents
`should be entering into early clinical trials in the
`near future.
`
`Arsenic Trioxide
`Arsenic was used as a medicinal 2,400 years ago
`in the time of the ancient Greeks and Romans.
`Paul Ehrlich used organic arsenicals for the treat-
`ment of syphilis. Arsenicals
`are still
`included as
`ingredients
`in folk remedies of some cultures,
`particularly
`in China
`and other parts of Asia.
`Arsenic was widely used to treat syphilis before the
`advent of penicillin,
`and the organic arsenical
`melarsoprol
`is a recognized treatment
`for the
`meningoencephalitic
`stage of African trypanoso-
`mtasis." Fowler's solution (1% arsenic trioxide in
`
`in the 18th
`formulated
`potassium bicarbonate),
`century to treat a variety of infectious and neoplas-
`tic disorders, was reported by US physicians in the
`1930s to be useful
`in the treatment
`of chronic
`myelogenous
`leukemia (CML), and more recently
`by hematologists in China to treat various forms of
`leukemia,
`including CML.97
`Recent
`interest in the development of arsenic
`trioxide as an anticancer agent emanates
`from
`reports by Chinese investigators98,99of its efficacy
`in the treatment of APL. These favorable results in
`APL were confirmed
`in the United
`States by
`investigators at Memorial Sloan-Kettering Cancer
`Center
`(MSKCC).IOO
`Preclinical
`studies have shown that human APL
`cells are very sensitive to the growth-inhibitorv
`and cytotoxic effects of arsenic trioxide.101,102 Sen-
`sitivity to arsenic trioxide in vitro has also been
`demonstrated against a variety of other tumor
`types, including those derived from myeloid leuke-
`mias other
`than PML,101 myelorna.l'"
`lymphoid
`leukemia and lymphoma,I04-107prostate cancer,lOS
`and a various other solid rumors.P?
`The mechanism of antitumor activity of arsenic
`trioxide is not understood, but it appears to depend
`to some degree on dose and tumor type. The
`principal
`target
`in APL cells is the promvelocvric
`leukemia gene-retinoic
`acid receptor-alpha
`fusion
`transcript
`(PML/RAR_lY)102,110 Arsenic
`trioxide
`causes the degradation
`of the PML/RAR-lY and
`wild-type PML proteins,
`thus inhibiting their af-
`feet on growth and differentiation.lOZ,111,llZ Unlike
`ATRA, arsenic trioxide does not downregulate
`wild-type RAR_lY.102 In NB4 cells, a human APL
`cell line with t05;17)
`and the PML/RAR-o: fusion
`protein,
`the effects of arsenic trioxide treatment
`are dose-dependentl'":
`induction of partial (nonter-
`mtnal) differentiation at relatively low concentra-
`tions
`(0.1 to 0.5 pmol/L) with predominantly
`apoprosis at higher concentrations
`(0.5 to 2 pmol!
`L). However,
`induction of apoptosis by arsenic
`trioxide involves mechanisms other than modula-
`tion of PML or PML/RAR-lY.lol
`For example,
`growth inhibition and apoptosis induced by arsenic
`trioxide has been associated with downregulation
`of bel-2 expression in APL and other myeloid cell
`lines.101,102 Arsenic trioxide is known to react with
`sulfhydryl
`(SH) groups and thus alter many differ-
`ent enzyme systems,
`including those that affect
`protein tyrosine phosphorylation.Uv-U" The antitu-
`mor properties of arsenic trioxide have been attrib-
`
`DR. REDDY’S LABS., INC. EX. 1029 PAGE 6
`
`

`

`566
`
`CHESON ET AL
`
`to effects on mitochon-
`in part,
`least
`uted, at
`dria.105,115,116 Studies with isolated systems suggest
`that arsenic trioxide induces apoptosls by directly
`influencing the mitochondrial permeability transi-
`tion pore, which can be inhibited
`by Bci-Z, an
`endogenous antagonist of permeability transition
`pore funcnoo.!" Mitochondrial
`transmembrane
`potential collapse has been demonstrated in malig-
`nant
`lymphocytic cells undergoing arsenic trioxide-
`induced apoptosis.t'v Cells become more sensitive
`to arsenic trioxide when combined with inhibitors
`of glutathione
`(GSH) synthesis,
`including ascorbic
`acid.105,1l7 Additional data suggest that
`induction
`of mitotic arrest and apoptosis by arsenic trioxide is
`to
`related to the binding of this trivalent arsenical
`two cysteine residues in tubulin,
`thus blocking the
`guanosine
`triphosphate
`(GTP) binding site and
`leading to disruption
`of microtubule
`formation
`during mitosis. us
`Most clinical experience with arsenic trioxide
`comes from trials conducted
`in China
`and the
`United States
`in patients with refractory or re-
`lapsed APL,98.]OO,1I9.120In a study by Shen et al,99
`arsenic trioxide was administered intravenously at
`a fixed dose of 10 mg/d, either
`alone or
`in
`combination with other chemotherapy. Nine of 10
`patients treated with arsenic alone and all five of
`the remaining patients treated with a combination
`regimen achieved
`complete
`hematologic
`remis-
`sions. Niu et aP20 recently reported the results of
`arsenic trioxide
`treatment
`in 58 patients with
`APL. Clinical complete remission was obtained in
`eight of 11 patients (72.7%) with newly diagnosed
`(85.1%) with relapsed
`APL and 40 of 47 patients
`disease, In a pilot study conducted at MSKCC in
`the United Stares.F? arsenic trioxide was adminis-
`tered intravenously
`over 2 to 4 hours at doses
`ranging from 0,06 to 0.2 mg/kg/d; the initial course
`of treatment was continued until
`the bone marrow
`was morphologically clear of leukemia. The me-
`dian duration of induction
`therapy was 33 days
`(range, 12 to 39 days), with one to five courses
`administered to responding patients. A complete
`hematologic
`remission was achieved in 11 of the
`12 patients. The median duration of remission was
`(range, 1 to >9 months),
`5 months
`In eight
`patients,
`the bone marrow tested negative
`by
`reverse-transcription
`polymerase
`chain
`reaction
`(RT PCR)
`for PML/RAR-Cl after
`two courses of
`therapy. The three patients whose bone marrow
`remained RT-PCR-positive
`relapsed during the
`
`second course of treatment. None of the patients
`whose bone marrows were RT~PCR-negative
`re-
`lapsed at the time of the report. The results of this
`single-institution
`study are being confirmed in a
`multicenter
`trial. 1Zl A dose-finding phase I study of
`arsenic trioxide is being conducted by investigators
`at MSKCC to determine the recommended
`phase
`II dose in hematologic malignancies
`other
`than
`APL,12l
`to arsenic trioxide
`attributed
`toxicities
`Major
`involve a variety of organ systems. Skin changes
`are common,
`including
`dryness, hyperkeratosis,
`pruritus,
`rash, erythema,
`and hyperpigmentanon.
`Gastrointestinal
`toxicity includes nausea, vomit-
`ing, abdominal
`pain,
`anorexia,
`and stomatitis.
`Renal and hepatic toxicity and hyperglycemia are
`also observed. Patients can experience a variety of
`constitutional
`symptoms
`and muscular
`skeletal
`complaints,
`such as lassitude, fatigue, weight gain,
`arthralgia,
`bone pain, myalgia,
`toothache,
`and
`headache. Cardiovascular
`and neurologic complt-
`cations may be a particular
`concern in patients
`who receive more than a few courses of therapv.!'?
`Neurotoxicity includes peripheral motor or sensory
`neuropathy and seizures. Some patients experience
`lightheadedness. dizziness, and hypotension during
`the 1~ or 2-hour
`infusion, but
`this
`is usually
`alleviated by extending
`the infusion to 4 hours.
`Other
`cardiac
`effects
`that have been reported
`include second-degree heart block, prolongation of
`the QT interval, and torsades des pointes; patients
`exposed to amphotericin B, or those with hvpoka-
`lemia or hypomagnesemia for any reason, may be at
`higher risk for these cardiac abnonualities.Uv
`Of interest
`is that patients with APL treated
`with arsenic trioxide can develop fluid retention
`and a syndrome
`similar
`to the
`"retinoic
`acid
`syndrome/'122,123with fever, fluid retention, weight
`gain, dyspnea, pneumonitis, and leukocytosis. Signs
`or symptoms of the syndrome are usually mani-
`fested within 1 to

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