throbber
Current role of thalidomide in cancer treatment
`Deborah A. Thomas, MD, and Hagop M. Kantarjian, MD
`
`Thalidomide (Thalidomid, Celgene, Warren, NJ) is a putative
`anti-angiogenesis and immunomodulatory agent that has
`demonstrated activity in various dermatologic and rheumato-
`logic conditions in addition to Crohn’s disease. The biologic
`effects of thalidomide and the clinical trials conducted in solid
`tumors, hematologic malignancies, chronic graft-versus-host
`disease (GVHD), and cancer-related cachexia are reviewed. A
`summary of the preliminary results of ongoing clinical trials is
`presented, and the future directions of thalidomide research in
`the oncology are discussed. Curr Opin Oncol 2000, 12:564–573
`© 2000 Lippincott Williams & Wilkins, Inc.
`
`University of Texas MD Anderson Cancer Center, Department of Leukemia,
`Houston, Texas, USA
`
`Correspondence to Deborah A. Thomas, MD, Assistant Professor, Department
`of Leukemia, University of Texas M. D. Anderson Cancer Center, 1515
`Holcombe Blvd, Box 61, Houston, TX 77030; e-mail: debthomas@mdander-
`son.org
`
`Current Opinion in Oncology 2000, 12:564–573
`
`Abbreviations:
`
`bFGF
`GM
`GVHD
`IL
`TNF-aa
`VEGF
`
`basic fibroblast growth factor
`glioblastoma multiforme
`graft-versus-host disease
`interleukin
`tumor necrosis factor alpha
`vascular endothelial growth factor
`
`ISSN 1040–8746 © 2000 Lippincott Williams & Wilkins, Inc.
`
`564
`
`Historical perspective
`Thalidomide was first introduced in 1953 by Chemie
`Grunenthal as an oral sedative-hypnotic [1]. It had no
`toxicity or addictive properties at therapeutic levels;
`however, peripheral neuritis was noted in some patients
`with long-term use. It was also used to ameliorate
`nausea and vomiting during pregnancy. In 1960, limb
`malformations and abnormalities of internal organs in
`newborns were associated with the maternal use of
`thalidomide. It was thus withdrawn from the market in
`1961 due to its teratogenecity [2].
`
`In 1965, the surprising activity of thalidomide in reac-
`tive lepromatous leprosy (seen during its use as a seda-
`tive-hypnotic) stimulated further study [3]. After a series
`of confirmatory placebo-controlled trials, thalidomide
`was approved by the Food and Drug Administration as
`treatment for the acute cutaneous manifestations of
`erythema nodosum leprosum and as maintenance
`therapy to prevent recurrence. Its immunomodulatory
`properties have been effective in other dermatologic
`diseases such as cutaneous lupus erythematosus, recur-
`rent erythema multiforme, and recurrent aphthous
`ulcers (especially in AIDS patients) [3–7].
`
`The first oncology studies of thalidomide were also
`reported in 1965. Grabstad and Golbey treated 71 patients
`with various malignancies with doses ranging from 300 mg
`to 2 g daily [8]. The one clinical responder had renal cell
`carcinoma metastatic to the lung and was treated after
`nephrectomy. Olson et al. [9] treated 21 patients with
`various solid tumors with doses ranging from 200 mg thrice
`daily up to 1400 mg daily. No tumor regressions were
`noted, but slowing of disease progression was observed in
`two patients, with palliation of symptoms in seven (33%).
`Because of the paucity of objective tumor responses, inter-
`est in thalidomide as an anti-cancer agent waned.
`
`Recently, investigators discovered that the biologic
`effects of thalidomide include inhibition of angiogenesis
`and suppression of tumor necrosis factor alpha (TNF-a).
`Owing to these unique mechanisms of action and favor-
`able side effect profile relative to chemotherapy (Table
`1), the study of thalidomide as a potential anti-tumor
`agent has intensified.
`
`Role for thalidomide in oncology:
`anti-angiogenesis
`In 1971, Folkman [10] determined that angiogenesis
`was essential for tumor development. Progressive
`
`DR. REDDY’S LABS., INC. EX. 1005 PAGE 1
`
`

`

`Current role of thalidomide in cancer treatment Thomas and Kantarjian 565
`
`Table 1. Rationale for thalidomide in oncology
`
`Parameter
`
`Onset at effect
`
`Chemotherapy
`
`Rapid
`
`Mechanism of action
`
`Specific to agent
`
`Thalidomide
`
`Slower
`
`Inhibits bFGF- and VEGF-angiogenesis
`Inhibits TNF-a
`, IL-12, IL-6
`Stimulates IL-2, IFN-g
`Stimulates CD8+ T cells
`Alters adhesion molecules
`
`Drug resistance
`
`Commonly acquired
`
`Less likely due to multiple targets without cross resistance
`
`Toxicity
`Alopecia
`Gastrointestinal
`Myelosuppression
`Wound healing
`Teratogenicity
`Secondary
`MDS/AML
`Neurologic
`
`Frequent
`Mucositis, nausea, diarrhea
`Frequent
`Delayed
`Known with specific agents
`Risk with some agents (eg,etoposide)
`Specific to agent (ara-C, ataxia; vincristine, peripheral
`neuropathy)
`
`Rare
`Constipation
`Rare, more common in HIV, GVHD
`Delayed
`Severe
`No known mutagenicity, carcinogenicity
`Somnolence; peripheral neuropathy with long-term use
`
`Clinical trial endpoint
`Phase I
`Phase II
`Combination
`
`Maximum tolerated dose
`Maximum biologic effect
`Tumor reduction
`Time to progression and survival
`Overlapping toxicities, reduced doses required for drug
`Synergy with chemotherapy, XRT, other
`combinations
`antiangiogenesis agents
`AML, acute myelogenous leukemia; bFGF, basic fibroblast growth factor; GVHD, graft-versus-host disease; IFN-g, interferon- g; IL-6, interleukin-6;
`IL-12, interleukin-12; MDS, myelodysplasia; TNF-a, tumor necrosis factor-alpha; VEGF, vascular endothelial growth factor; XRT, radiation therapy.
`
`recruitment of blood vessels to the tumor site resulted
`in proliferation and allowed cancer cells access to the
`vascular system for hematogenous spread. The degree
`of tumor vascularization is an independent prognostic
`factor for survival in carcinomas of the breast [11,12],
`lung [13], prostate [14,15], and esophagus [16]. The
`tumor vascular density also correlates with increased
`metastases, recurrences, or worse prognosis for carcino-
`mas of the bladder [17,18], colon [19], and stomach [20],
`and for melanoma [21].
`
`For hematologic malignancies, the pathologic correlate
`of tumor vascularization is bone marrow microvessel
`density. Studies in childhood acute lymphocytic
`leukemia [22], multiple myeloma [23–25], agnogenic
`myeloid metaplasia [26], and other acute and chronic
`leukemias [27] have shown increased marrow microves-
`sel density to correlate with poor risk features.
`Promoters of angiogenesis such as vascular endothelial
`growth factor (VEGF), basic fibroblast growth factor
`(bFGF), and TNF-a
`are elevated in several hematopoi-
`etic malignancies [28,29].
`
`In 1994, D’Amato et al. [30] showed that thalidomide
`inhibited bFGF-induced angiogenesis in a rabbit cornea
`micropocket assay. It also inhibited VEGF in a murine
`model of corneal vascularization [31]. In preclinical
`studies using a murine breast cancer model, thalidomide
`monotherapy did not reduce tumor size, but when
`combined with chemotherapy, prevention of distant
`
`lung and liver metastasis was observed compared with
`chemotherapy alone [32]. Other investigators have
`confirmed the anti-angiogenic effects of thalidomide
`after metabolic activation [33].
`
`Cytokine modulation and other biologic
`effects of thalidomide
`The level of TNF-a
`is elevated in various malignancies
`and other pathologic processes such as rheumatoid arthri-
`tis [34] and Crohn’s disease [34]. It enhances neo-angio-
`genesis, interacts with other proliferative cytokines such
`as interleukin (IL)-6, and contributes to many of the
`systemic symptoms of advanced malignancy. TNF-a
`can
`induce endothelial production and secretion of collage-
`nase, urokinase-type plasminogen activator, and plas-
`minogen activator inhibitor (PAI)-1 to promote tumor
`expansion and metastasis. It induces bFGF mRNA (and
`collagenase and IL-6 mRNA) in human omental
`microvascular cells [35]. Soluble receptors for TNF-a
`are
`present in human ovarian and breast carcinomas [36,37].
`
`Thalidomide inhibits TNF-a production by stimulated
`monocytes, macrophages, and neutrophils [38]. Proposed
`mechanisms include (1) decreased synthesis by acceler-
`ated degradation of TNF-a mRNA, (2) binding to a
`1-acid
`glycoproteins with intrinsic anti-TNF-a
`activity, and (3)
`decreased binding activity of NFkB, which controls the
`activation of the TNF-a gene [38–41]. Thalidomide also
`inhibits monocyte IL-12 production, enhances synthesis
`of IL-2, and inhibits IL-6 [42–44].
`
`DR. REDDY’S LABS., INC. EX. 1005 PAGE 2
`
`

`

`566 Therapeutic modalities
`
`Immunomodulatory effects on lymphocytes include (1)
`differential CD8+ T-cell stimulation resulting in
`decreased CD4/CD8 ratio, (2) shift from Th1 to Th2 T-
`cell responses, and (3) inhibition of T-cell proliferation
`in stimulated lymphocytes [45–48]. Thalidomide also
`inhibits neutrophil chemotaxis and modifies expression
`of surface adhesion molecules such as integrin receptors
`on leukocytes [49–51]. Direct anti-tumor effects of
`thalidomide include morphologic differentiation of the
`human leukemia cell line K562 in vitro [52].
`
`Pharmacology and toxicity
`Thalidomide is a racemate; the S and R isomer forms
`represent derivatives of l- and d-glutamic acid, respec-
`tively. The S isomer has been linked to teratogenicity,
`whereas the R isomer appears responsible for the seda-
`tive properties [53]. At physiologic pH, these isomers
`rapidly interconvert in vivo. The absolute bioavailability
`has not been characterized because of poor aqueous
`solubility. In studies of healthy volunteers and subjects
`with Hansen’s disease, the mean time to peak plasma
`concentrations ranged from 2.9 to 5.7 hours, indicating
`that it is slowly absorbed from the gastrointestinal tract
`[54••]. The influence of food on the rate or extent of
`absorption has not been determined.
`
`Side effects of thalidomide include somnolence,
`nausea, dry mouth and skin, skin rashes, constipation,
`increased appetite, headache, hypertension, bradycar-
`dia, dizziness and orthostatic hypotension, altered
`temperature sensitivity, irregularities in menstrual
`cycles, hypothyroidism, edema of lower extremities,
`teratogenicity, and peripheral neuropathy [54••].
`Constipation can be controlled with an aggressive laxa-
`tive regimen. Rash and leukopenia appear to be more
`common in AIDS patients and recipients of allogeneic
`bone marrow transplant. The immunomodulatory
`action of thalidomide has not been associated with an
`increased incidence of infections.
`
`Adverse effects such as somnolence, nausea, and skin
`rashes are dose-dependent and generally resolve with
`discontinuation of therapy. Tachyphylaxis has been
`observed to the somnolence and can be minimized by
`administration at bedtime. Thalidomide appears to
`enhance the sedative effects of barbiturates and alcohol
`and the catatonic effects of chlorpromazine and reser-
`pine. Thalidomide-induced sedation can be antagonized
`with the central nervous system stimulants
`methylphenidate and methylamphetamine.
`
`Thalidomide-associated neuropathy is generally charac-
`terized by painful symmetric paresthesias in the toes and
`feet, and electrophysiologic findings are often consistent
`with axonal degeneration without demyelination [55–57].
`Other associated symptoms include muscle cramps or
`
`weakness, signs of pyramidal tract involvement, and
`carpal tunnel syndrome. Rapid improvement is usually
`observed with discontinuation of therapy; however, cases
`of irreversible or longstanding sensory loss have been
`reported. The incidence of neuropathy appears to be
`increased in older patients and after administration of
`high cumulative doses; there may be a relation between
`slow acetylation and development of neuropathy.
`
`Thalidomide is teratogenic and should never be used by
`women who are pregnant or who could become pregnant
`while taking the drug. Major human fetal abnormalities
`have been documented: amelia (absence of limbs),
`phocomelia (short limbs), hypoplasticity of the bones,
`absence of bones, abnormalities or absence of organs
`(kidney, spleen, gallbladder), external ear abnormalities,
`facial palsies, eye abnormalities, and congenital heart
`defects. The highest risk occurs when the thalidomide is
`ingested during days 35 to 50 of pregnancy; severe effects
`can occur even with a single 100-mg dose. Thus, the drug
`must not be used at any time during pregnancy, and
`recommended contraceptive methods must be used by
`both women and men of child-bearing potential. Females
`on rifampin, rifabutin, barbiturates, steroids, phenytoin or
`carbamazepine should not rely on hormonal contracep-
`tion, since efficacy is reduced. Studies of the plasma phar-
`macokinetics of oral contraceptives given concomitantly
`with thalidomide do not indicate reduced efficacy [58].
`The S.T.E.P.S. program has been instituted to reduce
`fetal exposure; access is controlled by registration of
`prescribing physicians, dispensing pharmacies, and
`patients [59•]. A comprehensive consent process with
`educational materials is mandatory.
`
`Clinical trials in solid tumor malignancies
`High-grade gliomas
`Two early phase II studies have shown promising activ-
`ity of thalidomide in patients with recurrent high-grade
`gliomas (highly vascular tumors with high microvessel
`density are an adverse prognostic factor) such as
`anaplastic mixed glioma, anaplastic astrocytoma, or
`glioblastoma multiforme (GBM). Fine et al. [60••]
`administered 800 mg to 1200 mg daily to patients previ-
`ously treated with external radiation therapy with or
`without chemotherapy. Two of 36 patients (6%) had
`partial responses, two (6%) had minor responses, and 12
`(33%) had stable disease. Median time to progression
`was only 10 weeks for the entire group, but it was 33
`weeks for the responders, 15 for those with stable
`disease, and 8 weeks for the nonresponders. Median
`survivals were 74, 30, and 22 weeks, respectively.
`Increases in bFGF correlated with progression and
`shorter survival. Adverse events were grade 4 seizures in
`four patients with prior history of seizures or tumor
`progression. Somnolence and constipation were the
`main toxicities attributable to thalidomide.
`
`DR. REDDY’S LABS., INC. EX. 1005 PAGE 3
`
`

`

`Current role of thalidomide in cancer treatment Thomas and Kantarjian 567
`
`In another single-agent trial, 37 patients with recurrent
`GBM were treated with doses ranging from 100 mg to
`500 mg daily [61]. Of the 34 patients evaluable, 5 (15%)
`had a partial response, 11 (32%) had stable disease, and
`18 (53%) had progressive disease. No correlation
`between VEGF levels and outcome was observed. In a
`combination trial, 46 patients with recurrent high-grade
`gliomas were treated with carboplatin (AUC 8) and
`thalidomide 300 mg/m2 daily [62]. Five partial responses
`(12%) were observed and 28 (70%) patients had stable
`disease with a median response duration of 24 weeks
`and estimated survival of 40 weeks. Hematologic toxic-
`ity was prominent in patients receiving prior nitrosurea.
`Major nonhematologic toxicities attributable to thalido-
`mide were constipation and drowsiness. Complete
`results of these two trials are not yet published.
`
`Kaposi’s sarcoma
`Recent data indicate that thalidomide is active in AIDS-
`related Kaposi’s sarcoma. Fife et al. [63] conducted a
`phase II study of 100 mg daily for 8 weeks in 17 patients
`with cutaneous AIDS-related Kaposi’s sarcoma. Six of 17
`patients achieved a partial response (35%), and 8 patients
`withdrew from therapy because of toxicity (n=6),
`progression (n=1), or noncompliance (n=1). Interestingly,
`human herpesvirus 8 DNA load became undetectable in
`three of the five assessable partial responders.
`
`Preliminary results of two additional studies have been
`reported. Politi et al. [64] treated 12 patients with doses
`ranging from 200 to 600 mg daily. Two of 12 patients
`(17%) responded at doses of 200 and 400 mg per day,
`respectively; 7 patients (58%) had stable disease. The
`partial responses lasted for 3+ and 8 months. The
`median time to disease progression was 4 months. Dose-
`dependent somnolence was observed, requiring cessa-
`tion of therapy in two of three patients treated at 600 mg
`per day. Rash, headache, and paresthesias were
`observed. Yarchoan et al. [65] treated 13 patients with
`AIDS-related Kaposi’s sarcoma in an escalating fashion
`beginning with 200 mg up to 1000 mg as tolerated. Four
`of 11 evaluable patients (36%) achieved a partial
`response. Two maintained their response at 30+ and 50+
`weeks, whereas two progressed at 16 and 49 weeks. Five
`patients (45%) had stable disease ranging from 12 to 52
`weeks. Rash, neutropenia, depression, and sedation
`were reported.
`
`Renal cell carcinoma
`In a large phase II study, Eisen et al. [66•] treated 66
`advanced cancer (ovarian, melanoma, renal, breast)
`patients with thalidomide 100 mg daily. Three of 18
`patients (17%) with renal cell carcinoma had partial
`responses, and 3 had stable disease for 3 months or
`longer. Two of the responders had failed bioim-
`munochemotherapy; tumor shrinkage was observed
`
`within 2 weeks of starting thalidomide. No other objec-
`tive responses were observed, but patients had reduced
`insomnia and stabilization of weight loss. Toxicities
`included grade 2 lethargy (n=8), grade 2 neuropathy
`(n=2), and skin rashes. Progression correlated with
`increasing serum and urine levels of VEGF. Preliminary
`results of another phase II study in 15 patients with
`renal cell carcinoma used higher doses ranging from 400
`mg to 1200 mg daily [67]. Of the 12 patients evaluable
`for response (at least 4 weeks of therapy), 1 had a partial
`response for 11+ months after failing IL-2, 1 had a minor
`response for 3+ months, 3 had stable disease, and 7
`patients had progressive disease. Full results of this trial
`are not yet available.
`
`Prostate cancer
`Figg et al. [68] reported preliminary data of thalidomide
`in 63 patients with metastatic prostate cancer who had
`failed combined androgen blockade as well as antiandro-
`gen withdrawal. The drug was administered at either a
`low dose (n=50, 200 mg daily) or high dose (n=13, titra-
`tion to 1200 mg). Declines in prostate-specific antigen
`were observed in 58% and 68% of patients on the low-
`dose and high-dose arms, respectively. Eighteen percent
`treated with low-dose thalidomide had greater than 50%
`reductions in prostate-specific antigen, the longest
`maintained for 1.5 years. No objective tumor reductions
`were observed, but two patients had both symptomatic
`and radiographic improvement of bone scan lesions.
`Reduced bFGF levels were seen in responders. The
`most prevalent complaints were constipation, dizziness,
`edema, fatigue, xerostomia, and neurocortical symp-
`toms. Peripheral neuropathy was observed with long-
`term use (> 9 months). Thalidomide upregulates
`prostate-specific antigen secretion in the human
`prostate cell line LNCaP [69], indicating that additional
`end points may be required to determine response in
`future trials.
`
`Hepatocellular carcinoma
`Patt et al. [70] studied patients with nonresectable hepa-
`tocellular carcinoma beginning at 400 mg daily with esca-
`lation to 1000 mg by week 5 (one-third dose was adminis-
`tered in the afternoon and two-thirds at bedtime).
`Twenty-seven patients were treated; 21 were evaluable
`for response. Preliminary results included one partial
`response and one minor response; 10 patients (48%) had
`stable disease for at least 2 months. Somnolence was
`observed in all patients, and grade 3–4 skin reactions
`were observed in 20%. One patient developed an exfolia-
`tive dermatitis. Combination trials with chemotherapeu-
`tic agents such as capecitabine are planned.
`
`Melanoma
`Reported thalidomide experience is limited in
`melanoma. Eisen et al. [66•] treated 17 patients with
`
`DR. REDDY’S LABS., INC. EX. 1005 PAGE 4
`
`

`

`568 Therapeutic modalities
`
`advanced melanoma with low-dose thalidomide (100 mg
`daily). No objective responses were observed, but 4
`patients (24%) had stable disease for up to 5 months.
`One patient was noted to have significant symptom
`control of his leg deposits.
`
`Breast and ovarian cancer
`Eisen et al. [66•] treated 12 patients with advanced
`breast cancer and 19 with ovarian carcinoma with low-
`dose thalidomide (100 mg daily); no objective responses
`were observed in these groups. Nguyen et al. [32]
`treated advanced breast cancer patients (n=7) with
`thalidomide (100 to 300 mg daily) in combination with
`cyclophosphamide, doxorubicin, and fluorouracil (CAF).
`Three patients had partial response, one had stable
`disease, two had progressive disease, and one was lost to
`follow-up.
`
`Long et al. [71] treated seven patients with breast cancer
`after intensive chemotherapy and autologous stem cell
`transplant with thalidomide 400 mg orally daily begin-
`ning at hematologic recovery until day 180. Fatigue and
`mild somnolence were observed at the time of the
`report. Modulation of VEGF was noted. Full results of
`this study were not available; accrual was ongoing.
`
`Squamous cell carcinoma of the head and neck and
`non-small cell lung cancer
`Preliminary results of a phase II trial of escalating
`thalidomide (200 to 1200 mg) in 17 heavily pretreated
`patients with recurrent or metastatic squamous cell
`carcinoma of the head and neck was recently reported
`[72]. No objective tumor responses were observed, with
`94% of the patients discontinuing thalidomide due to
`progressive disease. The median survival of 5.4 months
`was similar to that of historical control subjects.
`
`A pilot study of thalidomide in combination with pacli-
`taxel 225 mg/m2 over 3 hours with carboplatin (AUC 6)
`was conducted in patients with advanced non-small cell
`lung cancer (unresectable stage IIIA, IIIB, or stage IV)
`[73]. Thalidomide was given 200 mg daily and escalated
`to 1000 mg as tolerated. Patients with stage III disease
`without pleural effusion received two cycles of combina-
`tion therapy followed by external radiation therapy plus
`thalidomide; those with a pleural effusion or stage IV
`disease received six cycles of combination therapy
`without external radiation therapy. Preliminary results
`in nine patients showed no objective responses; all six
`patients receiving two cycles of chemotherapy had
`stable disease. One stage IIIB patient with a pleural
`effusion had progressive disease. The regimen was well
`tolerated, with fatigue, myalgias, constipation, and grade
`I neuropathy the most common complaints. The mean
`tolerated dose of thalidomide was 711 mg. Accrual was
`ongoing at the time of the report.
`
`Miscellaneous solid tumor malignancies
`The Food and Drug Administration summarized the
`results of a physician survey regarding 575 single-
`patient Investigative New Drug applications issued for
`thalidomide in 1997 and 1998 [74]. Of the 544 practi-
`tioners surveyed, 359 (66%) responded to yield data on
`480 patients whose age ranged from 11 to 90 years
`(median 52 years). The most common malignancies
`treated were those of the breast, central nervous system
`(GBM), prostate, skin (melanoma), colon, pancreas, and
`kidneys (renal cell carcinoma). Thalidomide was given
`in doses up to 2400 mg daily, with 400 mg the most
`common dose (33%), followed by 200 mg (26%), 800
`mg (18%), and 1200 mg (15%). Fifty-eight patients
`(31%) were prescribed thalidomide in combination with
`chemotherapy. Responses were observed in 36 patients
`(7.5%), 10 of those with combination therapy. Most
`patients (53%) discontinued therapy for progressive
`disease, whereas only 10% ceased thalidomide because
`of toxicity. The most common side effects reported
`were somnolence, constipation, rash, fatigue, and
`mental status changes. Overall it appeared that thalido-
`mide was well tolerated with no increased toxicities
`attributable to combination therapy.
`
`Clinical trials in hematologic malignancies
`Multiple myeloma, plasma cell leukemia,
`Waldenstrom’s macroglobulinemia
`The remarkable efficacy of single-agent thalidomide in
`84 patients with refractory or relapsed multiple
`myeloma (90% relapsed after high-dose chemotherapy)
`was reported recently by Singhal et al. [75••]. The initial
`dose was 200 mg daily with escalation by 200 mg every 2
`weeks to a maximum dose of 800 mg. The maximum
`tolerable dose was then continued until progressive
`disease. Median duration of therapy was 52 days (range,
`2 to 465) with 80% receiving at least 4 weeks of therapy.
`
`Reduction in paraprotein levels (for at least 6 weeks) was
`observed in 32% of the patients. Two patients achieved a
`complete response. Responses in paraprotein correlated
`with reduction in plasma cells and improvements in
`hemoglobin levels and were apparent within 2 months in
`78% of the patients. Mild to moderate constipation,
`weakness, fatigue, or somnolence occurred in one third of
`the patients; 11% discontinued therapy due to intoler-
`ance. Neurologic side effects included somnolence, dizzi-
`ness, confusion, tremors, incoordination, tingling, and
`numbness. Decreases in marrow marrow microvessel
`density were observed; no direct correlation with
`response could be demonstrated. An update by Barlogie
`summarizing the Arkansas experience with thalidomide
`in multiple myeloma in over 300 patients (single-agent
`and combination therapy) confirmed the activity of
`thalidomide monotherapy in an additional 85 patients
`(total 169 patients) [76]. Eighty-four percent of the
`
`DR. REDDY’S LABS., INC. EX. 1005 PAGE 5
`
`

`

`Current role of thalidomide in cancer treatment Thomas and Kantarjian 569
`
`patients had some reduction in paraproteinemia (36% had
`‡25% reduction, 28%
`‡ 50% reduction, 20% ‡75% reduc-
`tion). Complete response or near-complete response was
`achieved in 12% of these patients. The activity of thalido-
`mide in refractory or relapsed multiple myeloma has been
`confirmed by other investigators [77,78].
`
`Combination therapy strategies are being explored in
`multiple myeloma. Weber et al. [79] reported on the effi-
`cacy of thalidomide in combination with pulse dexam-
`ethasone. Initially, 44 patients with relapsed or refractory
`multiple myeloma were treated with single-agent thalido-
`mide with an overall response rate of 25%. Nonresponders
`were treated with dexamethasone 20 mg/m2 for 4 days—
`days 1 through 4, 9 through 12, and 17 through 20—along
`with their maximum tolerated thalidomide dose. Four of
`10 patients (40%) with primary resistant disease and 5 of
`16 patients (31%) with resistant relapse achieved a partial
`response. The combination of thalidomide with
`chemotherapy (DT-PACE [dexamethasone, cisplatin,
`doxorubicin, cyclophosphamide, and etoposide]) has also
`proved an effective strategy for plasma cell leukemia and
`fulminant multiple myeloma [80•].
`
`Preliminary results in Waldenstrom’s macroglobulinemia
`have been reported [81]. Eight patients were treated with
`thalidomide at doses from 200 mg up to 600 mg daily;
`seven were evaluable for response. Results indicated that
`three patients had at least 50% reduction of serum mono-
`clonal protein, one had stable disease, and three had
`progressive disease. Reductions in organomegaly and
`splenomegaly were noted in responders.
`
`Myelodysplasia, acute leukemias, and chronic
`leukemias
`Raza et al. [82] reported preliminary results of thalido-
`mide in early myelodysplasia in 61 patients treated with
`100 to 400 mg of thalidomide. Of these, 22 had refractory
`anemia (RA), 13 had refractory anemia with ringed sider-
`oblasts, 19 had refractory anemia with excess blasts, 4 had
`refractory anemia with excess blasts in transformation,
`and 3 had chronic myelomonocytic leukemia. Of the 25
`patients evaluable for response, 17 (68%) responded and
`8 (32%) failed. Trilineage (n=3), bilineage (n=4), and
`monolineage (n=10) responses were observed. Criteria for
`responses were not clearly defined. Transfusion indepen-
`dence was reported, and some responses were not mani-
`fested until after 3 months of therapy.
`
`At the MD Anderson Cancer Center, thalidomide has
`been studied in relapsed or refractory myelodysplasia
`(refractory anemia with excess blasts, refractory anemia
`with excess blasts in transformation), acute myelogenous
`leukemia, chronic myelomonocytic leukemia, and chronic
`myelogenous leukemia [83,84]. Of the 14 patients with
`relapsed or refractory acute myelogenous leukemia, one
`
`had significant hematologic improvement (clearance of
`peripheral blasts, reduction in marrow leukemia infiltrate,
`and recovery of neutrophil count). Of the 10 patients with
`advanced myelodysplasia, one patient achieved a
`complete response. Transient reductions in marrow
`leukemic infiltrate were observed in three patients. Two
`of six patients with chronic myelomonocytic leukemia
`had hematologic improvements with reductions in
`splenomegaly and resolution of leukemia cutis. Of six
`patients with chronic myelogenous leukemia, one late
`chronic phase patient had a partial cytogenetic response;
`another had a complete hematologic response. All
`patients with chronic myelogenous leukemia–chronic
`phase had a reduction in requirements of concomitant
`cytoreductive agents such as hydroxyurea and anagrelide.
`Patients with rapidly progressive disease prior to institu-
`tion of thalidomide were unlikely to complete therapy.
`Toxicities included dose-limiting fatigue (59%),
`reversible neurotoxicity (46%), constipation (30%), and
`rash or skin dryness (19%). One third of the patients
`developed infections, usually occurring in the nonrespon-
`ders with prolonged neutropenia because of their under-
`lying disease. Randomized trials of anthracycline and ara-
`C induction chemotherapy with or without thalidomide
`are being conducted in poor-risk patients with acute
`myelogenous leukemia [83].
`
`Philadelphia-negative myeloproliferative disorders and
`myelofibrosis
`Preliminary results of thalidomide (escalating from 200
`mg to 800 mg) in 18 patients with either Ph-negative
`myeloproliferative disorders or myelofibrosis (primary or
`secondary) were recently reported [85]. Of twelve evalu-
`able patients, 8 (67%) responded with hematologic
`improvements in at least one of the following: (1) reduc-
`tion of splenomegaly or hepatomegaly by 50% or greater,
`(2) achievement of transfusion-independence and/or
`increase in hemoglobin by at least 2 g/dL, or (3) normal-
`ization of platelet count (> 100 · 109/L) with prior throm-
`bopenia. Most responders (88%) had improvements in at
`least two of the categories. Responses were gradual;
`reduction in organomegaly could take 3 months to
`achieve. Those with proliferative disease had a reduced
`need for concomitant cytoreductive agents. Grade 1 to 2
`toxicities included fatigue (72%), constipation (44%), skin
`rash (39%), and reversible neurologic difficulties such as
`incoordination, paresthesias, and tremors (28%). Therapy
`was discontinued in 39% of the patients for either toxicity
`(n=3) or progression (n=4). Continued study is planned.
`
`Supportive care
`Chronic graft-versus-host disease after allogeneic bone
`marrow transplant
`To determine whether thalidomide prophylaxis could
`prevent GVHD after allogeneic bone marrow transplant,
`a double-blind, randomized trial was conducted in 59
`
`DR. REDDY’S LABS., INC. EX. 1005 PAGE 6
`
`

`

`570 Therapeutic modalities
`
`patients [86]. Thalidomide was given in a dose of 200
`mg twice daily starting day 80. Surprisingly, a higher
`rate of GVHD was observed with thalidomide compared
`with placebo (82% vs 54%, respectively, P = 0.06) in
`addition to a higher mortality (39% vs 8%, P = 0.0006).
`
`istration, and continued for 3 months or 6 months in
`patients with complete response or partial response,
`respectively. Fourteen patients (32%) had a complete
`response, 12 (27%) had a partial response, and 18 (41%)
`failed. Toxicities were reported to be minimal.
`
`Vogelsang et al. [87] reported on 23 patients with chronic
`GVHD and 21 with high-risk chronic GVHD (defined
`as two of three risk factors: evolution from acute GVHD,
`lichenoid skin or mucous-membrane changes, or hepatic
`dysfunction). Thalidomide was given at a dose that
`produced a plasma level of 5 µg/mL 2 hours after admin-
`
`Parker et al. [88] studied 80 patients with thalidomide for
`patients with either progressive GVHD despite at least 1
`month of cyclosporin and/or prednisone (n=31), stable but
`persistent GVHD after at least 2 months of therapy (n=40),
`GVHD responsive to therapy but flaring after taper
`(requiring at least 20 mg prednisone, n=7), or medical
`
`Table 2. Examples of current or planned clinical trials of thalidomide in oncology
`
`Disease type
`
`Solid tumors
`Gastrointestinal
`Colorectal
`Biliary carcinoma
`Hepatocellular
`
`Eligibility
`
`Regimen
`
`Resected disease after recurrence
`Variable
`Variable
`
`Adjuvant vs. placebo
`Thalidomide
`Thalidomide + capecitabine
`
`SCCa head and neck
`
`Recurrent or metastatic disease
`
`Thalidomide
`
`Site(s)
`
`NCI
`MDACC
`MDACC
`
`MDACC
`
`Nonsmall cell lung
`
`Stage III
`
`Carboplatin, paclitaxel, XRT ± thalidomide
`
`NCI
`
`Prostate adenocarcinoma
`Androgen-independent
`
`Androgen-dependent
`
`Metastatic
`Metastatic
`Stage IV nonmetastatic; hormonal ablation
`
`Weekly docetaxel ± thalidomide
`Paclitaxel + estramustine + thalidomide
`Thalidomide vs. placebo
`
`Locally advanced
`
`Surgical candidate
`
`Pre-op thalidomide followed by RRP
`
`Ovarian epithelial
`
`Stage IC-IV
`
`Carboplatin ± thalidomide
`
`Renal cell carcinoma
`
`IL-2

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