throbber
C o m b i n a t i o n T h e r a p y W i t h T h a l i d o m i d e P l u s D e x a m e t h a s o n e
`f o r N e w l y D i a g n o s e d M y e l o m a
`
`By S. Vincent Rajkumar, Suzanne Hayman, Morie A. Gertz, Angela Dispenzieri, Martha Q. Lacy, Philip R. Greipp, Susan Geyer,
`Nancy Iturria, Rafael Fonseca, John A. Lust, Robert A. Kyle, and Thomas E. Witzig
`
`Purpose: Multiple myeloma is a malignancy of plasma
`cells and is characterized by increased marrow angiogene-
`sis. Thalidomide, an agent with antiangiogenic properties,
`is effective in relapsed myeloma. We report the results of a
`study combining thalidomide and dexamethasone as initial
`therapy for myeloma.
`Patients and Methods: Fifty patients with newly diag-
`nosed myeloma were studied. Thalidomide was given at a
`dose of 200 mg/d orally. Dexamethasone was given at a
`dose of 40 mg/d orally on days 1 to 4, 9 to 12, and 17 to 20
`(odd cycles) and 40 mg/d on days 1 to 4 (even cycles),
`repeated monthly.
`Results: Of all 50 patients, a confirmed response was
`seen in 32 patients yielding a response rate of 64% (95%
`confidence interval, 49% to 77%). Thirty-one patients (62%)
`proceeded to stem-cell collection after four cycles of therapy
`including 26 who underwent stem-cell transplantation and
`
`MULTIPLE MYELOMA accounts for 10% of malignant
`
`hematologic neoplasms.1,2 Recent evidence suggests that
`angiogenesis is increased in multiple myeloma and has prognos-
`tic value in this disease.3-5 On the basis of the increased
`angiogenesis observed in myeloma, thalidomide has been stud-
`ied as an antiangiogenic therapy. Although its mechanism of
`action in myeloma is unclear, several trials show that thalido-
`mide is active in 25% to 35% of patients with relapsed,
`refractory myeloma.6,7
`The current standard of care for patients with myeloma who
`are in good performance status is combination chemotherapy
`using non–alkylator-containing regimens such as vincristine,
`doxorubicin, and dexamethasone (VAD) for 4 to 6 months
`followed by high-dose therapy with autologous stem-cell trans-
`plantation. Such an approach has been shown to improve both
`response rates and survival.8-10 The response rate with VAD is
`approximately 55% to 65%.11 The goal of pretransplantation
`chemotherapy with VAD and similar regimens is to reduce
`tumor burden before stem-cell collection and transplantation.12
`VAD is preferred over regimens such as melphalan and pred-
`nisone and other alkylator-based regimens because it is less toxic
`to bone marrow stem cells, which is an important consideration for
`stem-cell transplant. However, VAD has significant toxicity and is
`cumbersome, requiring an indwelling central venous access for
`continuous infusion of chemotherapy that places the patient at risk
`for catheter-related infection, sepsis, and thrombosis.
`Dexamethasone has significant single-agent activity in my-
`eloma in both previously untreated and relapsed disease.13,14
`Recently, Weber et al15 demonstrated a clinical benefit by the
`addition of dexamethasone to patients for whom thalidomide
`alone was unsuccessful. On the basis of these results and the ease
`of administration over infusional VAD and other intravenous
`regimens, we conducted a clinical
`trial of thalidomide and
`dexamethasone in newly diagnosed myeloma. Because autolo-
`
`five who chose stem-cell cryopreservation. Major grade 3 or
`4 toxicities were observed in 16 patients (32%), and the
`most frequent were deep vein thrombosis (six patients),
`constipation (four patients), rash (three patients), and dys-
`pnea (two patients). Three deaths occurred during active
`therapy because of a pancreatitis, pulmonary embolism,
`and infection.
`Conclusion: We conclude that the combination of thalid-
`omide plus dexamethasone is a feasible and active regimen
`in the treatment of multiple myeloma. It merits further study
`as an oral alternative to infusional chemotherapy with
`vincristine, doxorubicin, and dexamethasone and other in-
`travenous regimens currently used as pretransplantation
`induction therapy for myeloma.
`J Clin Oncol 20:4319-4323. © 2002 by American
`Society of Clinical Oncology.
`
`gous stem-cell transplantation has been shown in randomized,
`controlled trials to be superior to conventional dose chemother-
`apy,
`the aim of the study was not
`to determine time to
`progression or survival with this regimen. Rather, our goal was
`to determine whether the combination of thalidomide and dexa-
`methasone would provide an orally administered, less toxic
`alternative to VAD (and other similar regimens) as pretransplan-
`tation induction therapy.
`
`PATIENTS AND METHODS
`
`Eligibility
`
`Patients were eligible to enter onto the study if they had previously
`untreated symptomatic myeloma. Patients were required to have bone
`marrow plasma cells ⱖ 10% and measurable disease defined as serum
`monoclonal (M) protein ⱖ 20 g/L and/or urine M protein ⱖ 400 mg/24
`hours. Patients with hemoglobin less than 70 g/L, platelets less than 25 ⫻
`less than 1,000 ⫻ 106/L, or Eastern
`109/L, absolute neutrophil count
`Cooperative Oncology Group (ECOG) performance score of 4 were ex-
`cluded. Pregnant or nursing women were not eligible. Women of child
`bearing potential who were unwilling to use a dual method of contraception
`and men who were unwilling to use a condom were not eligible for the study.
`All patients gave written informed consent before enrollment onto the study.
`
`From the Division of Hematology, Mayo Clinic and Mayo Foundation,
`Rochester, MN.
`Submitted February 23, 2001; accepted July 19, 2002.
`Supported in part by grant nos. CA85818, CA93842, and CA62242 from
`the National Cancer Institute, Bethesda, MD. S.V.R. is supported by the
`Multiple Myeloma Research Foundation and the Goldman Philanthropic
`Partnerships. S.V.R. and R.F. are also supported by Leukemia and Lym-
`phoma Society translational research awards.
`Address reprint requests to S. Vincent Rajkumar, MD, Division of
`Hematology, Mayo Clinic, 200 First St SW, Rochester, MN 55905; email:
`rajks@mayo.edu.
`
`Journal of Clinical Oncology, Vol 20, No 21 (November 1), 2002: pp 4319-4323
`DOI: 10.1200/JCO.2002.02.116
`
`4319
`
`DR. REDDY’S LABS., INC. EX. 1050 PAGE 1
`
`

`

`4320
`
`Approval of the study and consent form by the Mayo institutional review
`board was obtained in accordance with federal regulations and the Declara-
`tion of Helsinki. All physicians prescribing the drug and all study partici-
`pants adhered to the requirements of the System for Thalidomide Education
`and Prescribing Safety program. Women of childbearing age were required
`to have a pregnancy test performed every 2 weeks while on the study.
`
`Treatment Schedule
`
`Thalidomide was given orally at a dose of 200 mg/d for 2 weeks and then
`increased as tolerated by 200 mg/d every 2 weeks to a maximum dose of 800
`mg/d. After the first seven patients were enrolled, thalidomide dose escala-
`tion was discontinued because of unexpected skin toxicity in two patients,
`consisting of toxic epidermal necrolysis in one patient and generalized
`erythroderma in one patient.16 The dose of thalidomide was kept constant at
`200 mg/d for all subsequent patients. The thalidomide dose was reduced to
`50 to 100 mg/d if grade 2 or higher toxicity was encountered. Dexametha-
`sone was given at a dose of 40 mg/d orally on days 1 to 4, 9 to 12, and 17
`to 20 (odd cycles) and 40 mg/d days 1 to 4 (even cycles), repeated monthly.
`Patients were evaluated every 4 weeks for response. After four cycles of
`therapy, patients who were candidates for high-dose therapy were allowed to
`terminate study treatment to pursue stem-cell collection and transplantation.
`
`Response and Toxicity Criteria
`
`The primary end point of this trial was a confirmed response on two
`consecutive evaluations at least 4 weeks apart. The response and progression
`criteria used in this study are standard Mayo Clinic and ECOG criteria.
`Response was defined as a reduction of serum and urine M protein by at least
`50% accompanied by a similar reduction of soft tissue plasmacytomas if
`present. If response was assessed solely based on urine M protein, a 90% or
`greater reduction was required. In addition, responses were categorized as a
`complete response if there was complete disappearance of M protein in the
`serum and urine by immunofixation and absence of M plasma cells in the
`bone marrow. Disease progression was defined as a 50% increase in the M
`protein over the lowest response level. Increase in the size of existing lytic
`bony lesions or soft tissue plasmacytomas or the appearance of new lytic
`bony lesions constituted progression. A repeat M protein evaluation was
`required to confirm progression based on serum or urine monoclonal protein
`increase. However, as is standard in other myeloma protocols, if progression
`in M protein was accompanied by any other unequivocal evidence of
`progression, a repeat measurement was not necessary. Disease that does not
`satisfy the criteria for response, complete response, or progression was
`categorized as stable disease. The National Cancer Institute common toxicity
`criteria (version 2) were used to grade adverse effects.
`
`Statistical Analysis
`
`This trial was originally designed to accrue 30 patients to assess the
`response rate and toxicity of thalidomide and dexamethasone in the setting of
`newly diagnosed multiple myeloma. On the basis of promising activity noted
`at interim analysis, 20 additional patients were accrued to better define
`toxicity and response rate.
`Ninety-five percent confidence intervals (CIs) for the confirmed response
`probability were calculated using exact binomial 95% CIs. Toxicity inci-
`dence was estimated and summarized using frequency and descriptive
`techniques to assess any patterns. The Fisher’s exact test was used to
`compare differences in nominal variables.
`
`RESULTS
`
`Patient characteristics are listed in Table 1. The median age
`was 61 years (range, 33 to 78 years). The median serum M
`protein level before therapy was 3.8 g/dL (range, 0.0 to 7.6
`g/dL). The bone marrow plasma cell percentage before therapy
`ranged from 10% to 97% (median, 41%).
`Among the first seven patients treated, two had grade 3 or 4
`skin toxicity including one patient with toxic epidermal necroly-
`sis.16 One other patient had grade 2 exfoliation. Therefore, the
`protocol therapy was amended to stop the dose escalation of
`thalidomide and to keep the thalidomide dose constant at 200 mg
`for the subsequent 43 patients studied. Of the first seven patients,
`
`Table 1. Patient Characteristics
`
`RAJKUMAR ET AL
`
`All Patients
`
`Characteristic
`
`Sex
`Male
`Female
`Immunoglobulin heavy chain type
`IgG
`IgA
`Biclonal IgG and IgA
`Light-chain only (Bence Jones protein)
`Beta 2-microglobulin ⬎ 2.7 ␮g/mL
`Plasma cell labeling index ⱖ 1%
`Lactate dehydrogenase ⬎ 250 U/L
`Bone marrow plasma cell % ⬎ 40%
`
`No.
`
`31
`19
`
`33
`10
`1
`6
`31
`18
`5
`26
`
`%
`
`62
`38
`
`66
`20
`2
`12
`62
`36
`10
`52
`
`one patient did not receive dose escalation beyond 200 mg. The
`remaining six patients received a maximum thalidomide dose of
`400 mg. However, only two of these patients received this dose
`level for more than one cycle (one patient received 400 mg for
`six cycles and another for two cycles).
`
`Response to Therapy
`
`Thirty-two of the 50 patients (64%) had a response to therapy
`(95% CI, 49% to 77%). An additional 14 patients were catego-
`rized as stable disease and all had more than a 25% reduction in
`serum and urine M protein and can be considered as minor
`responses. If these minor responses are included, the overall
`response rate increases to 92%. The level of M protein reduction
`and bone marrow plasma cell reduction is listed in Table 2.
`When response was assessed by baseline M immunoglobulin
`(Ig) subtype, the confirmed response rates for IgG, IgA, and
`light-chain only were similar at 62%, 64%, and 60%, respec-
`tively. Four patients were taken off the study because of
`progressive disease after initial response.
`Response to therapy was accompanied by hematologic recov-
`ery and improvement in symptoms. Forty-seven patients had
`anemia at baseline (grade 1 in 29 patients, grade 2 in 16 patients,
`and grade 3 in two patients). Of these, 38% (18 of 47) patients
`had an increase in hemoglobin by at least 20 g/L or higher,
`
`Table 2.
`
`Extent of Monoclonal Protein and Bone Marrow Response
`
`Response Level
`
`No. of Patients
`
`Level of monoclonal protein reduction from baseline*
`90% or greater
`75%-90%
`50%-75%
`25%-50%
`No response or progression
`Level of bone marrow response from baseline†
`90% or greater
`75%-90%
`50%-75%
`25%-50%
`10%-25%
`No response or progression
`
`15
`11
`8
`12
`4
`
`10
`12
`6
`4
`4
`6
`
`%
`
`30
`22
`16
`24
`8
`
`24
`29
`14
`10
`10
`14
`
`*Urine monoclonal protein level was used when serum monoclonal protein level
`was less than 1 g/dL; two patients who achieved ⬎ 50% reduction in monoclonal
`protein levels in this Table were not considered responders because they did not meet
`all criteria for response and were classified as stable disease.
`†Of 42 patients in whom a repeat bone marrow biopsy was performed after
`protocol therapy.
`
`DR. REDDY’S LABS., INC. EX. 1050 PAGE 2
`
`

`

`THALIDOMIDE IN MYELOMA
`
`Table 3. Major (grade 3 and 4) Toxicities
`
`Type
`
`Treatment-related deaths
`Thrombosis
`Constipation
`Rash
`Dyspnea
`Depression
`Sedation
`Arrhythmia
`Rash
`Edema
`Inner ear
`Neuropathy
`Syncope
`
`No. of
`Patients
`
`3
`6
`4
`3
`2
`1
`1
`1
`1
`1
`1
`1
`1
`
`%
`
`6
`12
`8
`6
`4
`2
`2
`2
`2
`2
`2
`2
`2
`
`including both patients with grade 3 anemia and 11 of 16 patients
`with grade 2 anemia. One of the patients with improvement in
`anemia received erythropoietin support. Seven patients had
`baseline grade 1 or 2 leukopenia, which resolved with therapy in
`five patients. Eleven patients had baseline thrombocytopenia
`(grade 1 in nine patients, grade 2 in one patient, and grade 3 in
`one patient). Six of the nine patients with grade 1 thrombocyto-
`penia had an increase in platelet count by at least 50 ⫻ 109/L
`after therapy.
`Of the 50 patients treated, 31 proceeded to stem-cell collection
`after four cycles of therapy including 26 who underwent stem-
`cell transplantation and five who chose stem-cell cryopreserva-
`tion and are back on thalidomide plus dexamethasone. No
`problems were encountered with stem-cell collection. The re-
`maining 19 patients did not undergo stem-cell collection for
`varying reasons; this includes eight patients not considered to be
`transplant candidates because of age or patient refusal and were
`continued on thalidomide/dexamethasone therapy, four who
`proceeded to other therapy because of disease progression, four
`who pursued other therapy because of lack of adequate response
`or toxicity, and three patients who died while receiving therapy.
`
`Toxicity
`Therapy was generally well tolerated except for unexpected
`grade 3 or 4 skin toxicity in two of the first seven patients who
`had thalidomide dose escalation. There were three deaths that
`occurred during study treatment. One patient died with acute
`pancreatitis within 1 week of initiating therapy, which was
`attributed to either gallstones or dexamethasone therapy. One
`patient died within days of initiating therapy with a clinical
`diagnosis of pulmonary embolism, and one other patient died of
`infectious complications also in the first month of therapy. Grade
`3 or higher nonhematologic toxicity was seen in 17 patients
`(34%), and the most frequent were venous thrombosis in six
`patients (12%), constipation in four patients (8%), rash in
`three patients (6%), and dyspnea in two patients (4%). Table
`3 summarizes the grade 3 and 4 toxicities seen in the trial. The
`most common grade 1 and 2 toxicities were constipation
`(72%), neuropathy (58%), fatigue (50%), sedation (46%),
`rash (38%), tremor (30%), edema (28%), and elevated alka-
`line phosphatase (22%).
`
`DISCUSSION
`Thalidomide was first introduced in clinical practice as a
`sedative in the late 1950s and was subsequently withdrawn from
`
`4321
`
`the market in 1962 because of its severe teratogenicity. The
`mechanism of its teratogenicity is still unclear but may be related
`to its antiangiogenic properties or inhibition of tumor necrosis
`factor alpha (TNF ␣) production.17 Free radical mediated oxi-
`dative damage to DNA has also been postulated as a mechanism
`for the teratogenic effects.18 Despite its tragic past, thalidomide
`has re-entered clinical practice because of its immunomodula-
`tory and antiangiogenic properties. It was found to be effective
`in the treatment of erythema nodosum leprosum in the mid-
`1960s.19 In the last 10 years, thalidomide has been studied and
`found to be useful in the treatment of AIDS-related cachexia,
`aphthous ulcers in patients with Behc¸et’s disease, and in the
`treatment of chronic graft versus host disease.
`Clinical trials with thalidomide in myeloma were initiated
`because of its antiangiogenic properties and based on evidence
`indicating a role for increased angiogenesis in the pathogenesis
`and progression of myeloma.3,20 Researchers at the University of
`Arkansas conducted a landmark trial investigating the activity of
`thalidomide in relapsed myeloma.6 For most patients (90%) in
`this study, stem-cell transplantation had failed. Treatment con-
`sisted of thalidomide given at a dose of 200 mg/d orally for 2
`weeks and then increased by 200 mg/d every 2 weeks up to a
`maximum daily dose of 800 mg/d depending on toxicity. They
`observed an overall response rate of 32% with a median time to
`response of 1 month. M protein responses were accompanied by
`improvements in anemia and disease-related symptoms. The
`median duration of response was not reached after 14.5 months
`of follow-up. Considering that for 90% of the patients transplan-
`tation had failed, these results were impressive. An update to this
`study on 169 patients confirmed these results, demonstrating a
`2-year overall and event-free rates of 48% and 20%, respective-
`ly.21 Several groups including ours have since confirmed the
`activity of thalidomide in relapsed, refractory myeloma.7,22,23 A
`recent Mayo Clinic trial demonstrated a 38% response rate with
`the use of thalidomide in patients with previously untreated
`indolent or smoldering myeloma, indicating that thalidomide
`may have a role in early-stage disease as well.24
`The present study enrolled patients with newly diagnosed
`active myeloma, and most patients had high-risk disease. Sixty
`percent of those studied had high ␤2M levels, and 36% had a
`high plasma cell labeling index, which are two known adverse
`prognostic factors in myeloma. Only patients with marked
`cytopenias (hemoglobin ⬍ 70 g/L, platelet count ⬍ 25 ⫻ 109/L,
`or absolute neutrophil count ⬍ 1,000 ⫻ 106/L) were excluded
`from this study. The restrictions on cytopenias were placed
`because of safety concerns and concern that thalidomide may
`cause neutropenia. Further, when the study was designed, the
`combination had not been tested in newly diagnosed myeloma,
`and these restrictions excluded only a small minority of patients
`with myeloma. The study demonstrates significant activity (64%
`partial response rate) for the combination of thalidomide and
`dexamethasone in these patients. In addition, both the original
`Arkansas study in relapsed myeloma6 and the recent Mayo
`Clinic study in smoldering myeloma24 used less stringent criteria
`for measuring response (⬎ 25% reduction in serum and urine M
`protein) and observed response rates of 32% and 69%, respec-
`tively, with single-agent thalidomide. When this definition of
`response is used in the present study, the response rate with
`thalidomide plus dexamethasone increases to 92%. Thus, this
`combination merits further study as a simple oral alternative to
`
`DR. REDDY’S LABS., INC. EX. 1050 PAGE 3
`
`

`

`4322
`
`VAD as induction therapy in preparation for stem-cell transplan-
`tation. The toxicity seems lower, and the response rate is as good
`or better than that obtained using complex combination chemo-
`therapy regimens.11,12 None of the known prognostic markers in
`myeloma predicted response to therapy. More research is needed
`to determine which factors predict response to thalidomide
`therapy.
`This trial allowed patients to go off the study at 4 months to
`pursue stem-cell collection and transplantation. We believe that
`the current standard of therapy for multiple myeloma includes
`autologous stem-cell transplantation after three to four cycles of
`induction therapy with VAD or dexamethasone or a similar
`regimen. Randomized trials have shown this approach to be
`superior to conventional chemotherapy in terms of overall
`survival.25 In the United States, Medicare now covers autologous
`transplantation for newly diagnosed myeloma up to the age of
`78. The effect of prolonged thalidomide therapy on the collection
`of stem cells is unknown. For these reasons, we felt it was
`unethical to deny patients the option of stem-cell collection and
`transplantation. Thus, the study did not attempt to answer the
`question of time to progression with thalidomide and dexameth-
`asone therapy. The main goal of this study was to develop an
`alternative pretransplantation induction regimen for VAD that
`would be easier to use and possibly less toxic. However, a study
`comparing long-term thalidomide plus dexamethasone versus
`transplantation would be of interest in the future.
`The appropriate dose of thalidomide in myeloma is still
`unknown and is the subject of much debate. This trial was the
`first to combine thalidomide with dexamethasone for newly
`diagnosed myeloma. At the time this trial was designed, shortly
`after reports of the efficacy of thalidomide in relapsed myeloma,
`200 mg was felt to be the appropriate starting dose. During this
`study we learned that thalidomide doses more than 200 mg in
`combination with high-dose dexamethasone might be toxic.
`Doses less than 200 mg are being studied by other investigators
`in an attempt to minimize toxicity. In fact, grade 3 and 4
`neuropathy, constipation, and fatigue were infrequently seen in
`this study probably as a result of the relatively short duration of
`therapy for most patients, as well as the lower dose of thalido-
`mide compared with earlier studies in relapsed myeloma. Other
`studies have also found that toxicities are much lower with the
`200 mg dose of thalidomide compared with higher doses.6
`However, there is no data at present on whether there is any
`accompanying loss of efficacy.
`Most side effects can be managed by decreasing the dose of
`thalidomide to 50 to 100 mg/d. Because of our experience
`with skin toxicity at higher doses,16 we do not recommend
`escalating the dose of thalidomide beyond 200 mg when
`combining with dexamethasone. The increased skin toxicity
`seen with this combination is felt to be the result of potenti-
`ation of thalidomide toxicity by dexamethasone, the mecha-
`nisms for which are unclear.16 We also recommend that
`physicians exercise caution when combining thalidomide with
`other drugs known to cause skin toxicity, such as tri-
`methoprim-sulfamethoxazole and allopurinol.
`Twelve percent of patients had deep vein thrombosis (DVT) in
`this study. However, it is known that up to 10% to 20% of
`patients with newly diagnosed myeloma develop DVT in the first
`6 months of therapy.12 There is evidence that the combination of
`thalidomide with doxorubicin containing multiagent chemother-
`
`RAJKUMAR ET AL
`
`apy may increase the incidence of DVT.26,27 The incidence of
`DVT with single-agent thalidomide is less than 5%.21 It is not
`clear if the risk of DVT with thalidomide therapy is increased in
`the absence of doxorubicin and other cytotoxic agents.
`Most patients (52%) in this study will proceed to stem-cell
`collection and transplantation after four cycles of thalidomide
`and dexamethasone. In addition, some patients (10%) have
`elected to resume thalidomide and dexamethasone after stem-
`cell mobilization has been completed, opting to delay transplan-
`tation until disease progression. In this case, their stem cells are
`cryopreserved for future use. We currently mobilize stem cells
`after four cycles of therapy because the effects of prolonged
`thalidomide therapy on stem-cell yield is not known. Only eight
`patients not considered candidates for transplantation because of
`advanced age or poor performance status are continuing long-
`term thalidomide/dexamethasone therapy.
`The high response rate seen suggests a synergy between
`thalidomide and dexamethasone. There are in vitro data that lend
`support to this hypothesis.28 The mechanisms of the synergy are
`likely complex because both drugs have a wide variety of
`preclinical and clinical effects. One hypothesis is that dexameth-
`asone has a preferentially greater effect on the myeloma cells
`directly, and thalidomide effects are indirect, mediated through
`its effect on the microenvironment. It is also hypothesized that
`thalidomide helps in overcoming dexamethasone-induced resis-
`tance by its effects on adhesion molecules, tumor cell growth,
`and survival.28 In addition, both agents probably inhibit angio-
`genesis through different pathways.29,30 Preliminary results from
`Weber et al15 demonstrate responses with combined thalidomide
`and dexamethasone among patients with relapsed myeloma for
`whom previous treatment with the same agents given individu-
`ally had failed. The increased toxicity seen in this trial at higher
`doses of thalidomide in the first seven patients also supports a
`potentiation of thalidomide effects by dexamethasone.
`The mechanism of action of thalidomide in myeloma is
`unclear. Laboratory studies using the rabbit cornea micropocket
`assay have shown that thalidomide has potent antiangiogenic
`properties.30,31 Animal studies show that it can decrease vascular
`density in granulation tissue.32 In the Arkansas study, there were
`no statistically significant differences in posttreatment microves-
`sel density change between responders and nonresponders.6
`However, these findings do not fully exclude an antiangiogenic
`mechanism. In addition to its antiangiogenic effects, thalidomide
`also has several immunomodulatory properties. It inhibits the
`production of TNF␣ by enhancing the degradation of TNF␣
`mRNA.33 Thalidomide stimulates cytotoxic T-cell proliferation
`and induces the secretion of interferon gamma and interleukin-2
`by these cells.34 It may also modulate the expression of several
`cell surface adhesion molecules.35 Another potentially signifi-
`cant mechanism of action of thalidomide may be through
`inhibition of the transcription factor nuclear factor-kappaB.36,37
`The response rate seen in this study is higher than historical
`data with dexamethasone alone (43%) or thalidomide alone
`(38%) for previously untreated myeloma. Nevertheless,
`the
`results of this trial do not establish that thalidomide plus
`dexamethasone has superior response rate to dexamethasone
`alone. The response criteria used in this trial differ from the
`response criteria used in clinical
`trials using single-agent
`dexamethasone and other induction regimens used before
`tandem transplantation at the University of Arkansas. We
`
`DR. REDDY’S LABS., INC. EX. 1050 PAGE 4
`
`

`

`THALIDOMIDE IN MYELOMA
`
`4323
`
`believe that a randomized clinical trial comparing the two
`regimens is needed. Such a trial,
`led by ECOG,
`is now
`ongoing in the United States.
`We conclude that the response rate to induction therapy with
`the combination of thalidomide plus dexamethasone is similar to
`
`that expected with VAD and dexamethasone. Further studies are
`needed to document the effect on long-term outcomes posttrans-
`plantation with this regimen. Another strategy that needs study is
`to initiate induction therapy with dexamethasone alone and add
`thalidomide to patients who fail to respond.
`
`REFERENCES
`Identification of prognostic factors in a phase 2 study of 169 patients. Blood
`1. Greenlee RT, Murray T, Bolden S, et al: Cancer statistics, 2000. CA
`98:492-494, 2001
`Cancer J Clin 50:7-33, 2000
`22. Kneller A, Raanani P, Hardan I, et al: Therapy with thalidomide in
`2. Bataille R, Harousseau JL: Multiple myeloma. N Engl J Med 336:
`refractory multiple myeloma: The revival of an old drug. Br J Haematol
`1657-1664, 1997
`108:391-393, 2000
`3. Vacca A, Ribatti D, Roncali L, et al: Bone marrow angiogenesis and
`23. Juliusson G, Celsing F, Turesson I, et al: Frequent good partial
`progression in multiple myeloma. Br J Haematol 87:503-508, 1994
`remissions from thalidomide including best response ever in patients with
`4. Rajkumar SV, Leong T, Roche PC, et al: Prognostic value of bone
`advanced refractory and relapsed myeloma. Br J Haematol 109:89-96, 2000
`marrow angiogenesis in multiple myeloma. Clin Cancer Res 6:3111-3116,
`24. Rajkumar SV, Dispenzieri A, Fonseca R, et al: Thalidomide for
`2000
`previously untreated indolent or smoldering multiple myeloma. Leukemia
`5. Rajkumar SV, Witzig TE: A review of angiogenesis and anti-angio-
`15:1274-1276, 2001
`genic therapy with thalidomide in multiple myeloma. Cancer Treat Rev
`25. Attal M, Harousseau JL, Stoppa AM, et al: A prospective, randomized
`26:351-362, 2000
`trial of autologous bone marrow transplantation and chemotherapy in
`6. Singhal S, Mehta J, Desikan R, et al: Antitumor activity of thalidomide
`multiple myeloma: Intergroupe Francais du Myelome. N Engl J Med
`in refractory multiple myeloma. N Engl J Med 341:1565-1571, 1999
`335:91-97, 1996
`7. Rajkumar SV, Fonseca R, Dispenzieri A, et al: Thalidomide in the
`26. Zangari M, Anaissie E, Barlogie B, et al: Increased risk of deep-vein
`treatment of relapsed multiple myeloma. Mayo Clin Proc 75:897-902, 2000
`thrombosis in patients with multiple myeloma receiving thalidomide and
`8. Harousseau JL, Attal M: The role of autologous hematopoietic stem
`chemotherapy. Blood 98:1614-1615, 2001
`cell transplantation in multiple myeloma. Semin Hematol 34:61-66, 1997
`27. Osman K, Comenzo R, Rajkumar SV: Deep vein thrombosis and
`9. Barlogie B, Jagannath S, Epstein J, et al: Biology and therapy of
`thalidomide therapy for multiple myeloma. N Engl J Med 344:1951-1952,
`multiple myeloma in 1996. Semin Hematol 34:67-72, 1997
`2001
`10. Gertz MA, Pineda AA, Chen MG, et al: Refractory and relapsing
`28. Hideshima T, Chauhan D, Shima Y, et al: Thalidomide and its analogs
`multiple myeloma treated by blood stem cell transplantation. Am J Med Sci
`overcome drug resistance of multiple myeloma cells to conventional therapy.
`309:152-161, 1995
`Blood 96:2943-2950, 2000
`11. Alexanian R, Barlogie B, Tucker S: VAD-based regimens as primary
`29. Lansink M, Koolwijk P, van Hinsbergh V, et al: Effect of steroid
`treatment for multiple myeloma. Am J Hematol 33:86-89, 1990
`hormones and retinoids on the formation of capillary-like tubular structures
`12. Barlogie B, Jagannath S, Desikan KR, et al: Total therapy with
`of human microvascular endothelial cells in fibrin matrices is related to
`tandem transplants for newly diagnosed multiple myeloma. Blood 93:55-65,
`urokinase expression. Blood 92:927-938, 1998
`1999
`30. D’Amato RJ, Loughnan MS, Flynn E, et al: Thalidomide is an
`13. Alexanian R, Dimopoulos MA, Delasalle K, et al: Primary dexameth-
`inhibitor of angiogenesis. Proc Natl Acad Sci U S A91:4082-4085, 1994
`asone treatment of multiple myeloma. Blood 80:887-890, 1992
`31. Kenyon BM, Browne F, D’Amato RJ: Effects of thalidomide and
`14. Alexanian R, Barlogie B, Dixon D: High-dose glucocorticoid treat-
`related metabolites in a mouse corneal model of neovascularization. Exp Eye
`ment of resistant myeloma. Ann Intern Med 105:8-11, 1986
`Res 64:971-978, 1997
`15. Weber DM, Gavino M, Delasalle K, et al: Thalidomide alone or with
`32. Or R, Feferman R, Shoshan S: Thalidomide reduces vascular density
`dexamethasone for multiple myeloma. Blood 94:604a, 1999 (suppl 1, abstr)
`in granulation tissue of subcutaneously implanted polyvinyl alcohol sponges
`16. Rajkumar SV, Gertz MA, Witzig TE: Life-threatening toxic epider-
`in guinea pigs. Exp Hematol 26:217-221, 1998
`mal necrolysis with thalidomide therapy for myeloma. N Engl J Med
`33. Moreira AL, Sampaio EP, Zmuidzinas A, et al: Thalidomide exerts its
`343:972-973, 2000
`inhibitory action on tumor necrosis factor alpha by enhancing mRNA
`17. Argiles JM, Carbo N, Lopez-Soriano FJ: Was tumour necrosis
`degradation. J Exp Med 177:1675-1680, 1993
`factor-alpha responsible for the fetal malformations associated with thalid-
`34. Haslett PA, Corral LG, Albert M, et al: Thalidomide costimulates
`omide in the early 1960s? Med Hypotheses 50:313-318, 1998
`primary human T lymphocytes, preferentially inducing proliferation, cyto-
`18. Parman T, Wiley MJ, Wells PG: Free radical-mediated oxidative
`kine production, and cytotoxic responses in the CD8⫹ subset. J Exp Med
`DNA damage in the mechanism of thalidomide teratogenicity. Nat Med
`187:1885-1892, 1998
`5:582-585, 1999
`35. Geitz H, Handt S, Zwingenberger K: Thalidomide selectively modu-
`19. Iyer CG, Languillon J, Ramanujam K, et al: WHO co-ordinated
`lates the density of cell surface molecules involved in the adhesion cascade.
`short-term double-blind trial with thalidomide in the treatment of acute lepra
`Immunopharmacology 31:213-221, 1996
`reactions in male lepromatous patients. B

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