throbber
12 Dec 2001 8:13
`
`AR
`
`AR149-34.tex AR149-34.SGM
`
`LaTeX2e(2001/05/10)
`
`P1: GJC
`
`Annu. Rev. Med. 2002. 53:629–57
`
`THALIDOMIDE: Emerging Role in Cancer Medicine
`
`Paul Richardson, Teru Hideshima, and Kenneth Anderson
`Jerome Lipper Myeloma Center, Division of Hematologic Oncology, Department of
`Adult Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston,
`Massachusetts 02115; e-mail: paul richardson@dfci.harvard.edu
`
`Key Words
`immunomodulation, antiangiogenesis, myeloma
`n Abstract Thalidomide—removed from widespread clinical use by 1962 because
`of severe teratogenicity—has antiangiogenic and immunomodulatory effects, includ-
`ing the inhibition of tumor necrosis alpha factor. It has now returned to practice as
`an effective oral agent in the management of various disease states including ery-
`thema nodosum leprosum, for which it was approved by the U.S. Food and Drug
`Administration in 1998, and more recently certain malignancies, including multiple
`myeloma. Although thalidomide’s mechanism of action remains incompletely under-
`stood, considerable insight has been generated by extensive preclinical studies in mul-
`tiple myeloma. Moreover, clinical trials have confirmed benefit in relapsed disease, and
`the role of thalidomide in treating newly diagnosed patients is currently under study. Its
`use in other tumors is under evaluation, with promise in renal cell carcinoma, prostate
`cancer, glioma, and Kaposi’s sarcoma. Activity has also been demonstrated in chronic
`graft-versus-host disease and in symptom relief as part of palliative care.
`
`INTRODUCTION
`
`A tragic event in the history of drug development occurred with the over-the-
`counter marketing of thalidomide in Europe during the late 1950s for the treat-
`ment of pregnancy-associated morning sickness. As early as 1961, reports of
`teratogenicity and dysmyelia (stunted limb growth) associated with thalidomide
`use prompted its subsequent withdrawal (1, 2). The return of thalidomide as a
`therapy in certain conditions stems from its broad array of pharmacoimmuno-
`logic effects (3). This rehabilitation was reflected by its approval in 1998 by the
`U.S. Food and Drug Administration for the short-term treatment of cutaneous
`manifestations of moderate to severe erythema nodosum leprosum (ENL), along
`with its use as maintenance therapy to prevent and suppress the cutaneous man-
`ifestations of ENL recurrence (3). Thalidomide has since became a treatment of
`choice for ENL, and its wide spectrum of activity has fostered its application
`in a variety of disease states (Table 1) (3–5). Because of its teratogenic effects,
`thalidomide is now used under strict guidelines to prevent fetal exposure to the
`drug (4).
`
`629
`
`DR. REDDY’S LABS., INC. EX. 1043 PAGE 1
`
`

`

`12 Dec 2001 8:13
`
`AR
`
`AR149-34.tex AR149-34.SGM
`
`LaTeX2e(2001/05/10)
`
`P1: GJC
`
`630
`
`RICHARDSON ¥ HIDESHIMA ¥ ANDERSON
`
`TABLE 1 Potential therapeutic uses of thalidomide currently under investigation
`
`Cancer and related conditions
`
`Infectious diseases
`
`Autoimmune diseases
`
`Dermatologic diseases
`Other disorders
`
`Solid tumors (e.g., brain, breast, renal cell carcinoma);
`hematologic malignancies (e.g., multiple myeloma)
`HIV/AIDS and related conditions;
`aphthous ulcerations; wasting syndrome;
`mycobacterial infections (e.g., tuberculosis)
`Discoid and systemic lupus erythematosus;
`chronic graft-versus-host disease; inflammatory
`bowel disease; rheumatoid arthritis; multiple sclerosis
`Behcet’s syndrome; prurigo nodularis; pyoderma gangrenosum
`Sarcoidosis; diabetic retinopathy; macular degeneration
`
`In the field of medical oncology, the discovery of thalidomide’s antiangiogenic
`properties has coincided with the emerging importance of angiogenesis in tumor
`growth and progression. Thalidomide has been shown to inhibit angiogenesis in-
`duced by basic fibroblast growth factor (fl-FGF) in a rabbit cornea micropocket as-
`say and by vascular endothelial growth factor (VEGF) in a murine model of corneal
`vascularization (6, 7). In human studies, the drug appears to undergo activation
`to metabolites with antiangiogenic activity (8). Because of these antiangiogenic
`properties, thalidomide is currently undergoing evaluation in the treatment of var-
`ious solid tumors, multiple myeloma, and other hematologic malignancies (9–13).
`Results in multiple myeloma are particularly promising, although thalidomide’s
`antiangiogenic effects are believed to be only part of its antimyeloma activity.
`Its other potential actions include modulation of adhesion molecules, inhibition
`of tumor necrosis alpha factor (TNF-fi), downregulation of lymphocyte surface
`molecules, lowering of CD4:CD8 peripheral lymphocyte ratios, and direct effects
`on myeloma cells themselves (10, 14–18).
`This chapter presents a comprehensive review of the pharmacology of thalido-
`mide, a description of preclinical studies in multiple myeloma to illustrate the
`drug’s complex putative mechanisms of action, and a description of clinical stud-
`ies in multiple myeloma. Studies in other hematologic malignancies are also ad-
`dressed, as is the status of research in solid tumors and in other cancer-related
`applications.
`
`PHARMACOLOGY
`
`Thalidomide is a derivative of glutamic acid and is pharmacologically classi-
`fied as an immunomodulatory agent (19). Structurally, thalidomide contains two
`amide rings and a single chiral center (Figure 1); its full chemical name is alpha-
`Nfphthalimidogglutarimide [C13 O4 N2 H9] and its gram molecular weight is
`258.2 (19).
`
`DR. REDDY’S LABS., INC. EX. 1043 PAGE 2
`
`

`

`12 Dec 2001 8:13
`
`AR
`
`AR149-34.tex AR149-34.SGM
`
`LaTeX2e(2001/05/10)
`
`P1: GJC
`
`THALIDOMIDE
`
`631
`
`Figure 1 Structures of thalidomide and its potent ana-
`logues, immunomodulatory drugs (IMiDs).
`
`The currently available formulation is a nonpolar racemic mixture present as
`the optically active S and R isomers at physiologic pH, which can effectively
`cross cell membranes (19, 20). The S isomer has been linked to thalidomide’s
`teratogenic effects, whereas the R isomer appears to be primarily responsible for
`its sedative properties (7, 20, 21). The isomers rapidly interconvert at physiologic
`pH in vivo, and thus efforts at formulating only the R isomer have failed to obviate
`the teratogenic potential of thalidomide (20, 22).
`
`Pharmacokinetics
`Pharmacokinetic analysis of thalidomide in humans has been limited by the ab-
`sence of a suitable intravenous formulation, owing to the drug’s instability and
`poor solubility in water. The pharmacokinetics of thalidomide have therefore been
`determined only from animal studies and in humans receiving the oral therapy.
`Single-dose thalidomide trials have been conducted in healthy volunteers, patients
`with HIV infection, and patients with hormone-refractory prostate cancer (23–27).
`As Table 2 shows, the pharmacokinetics appear highly variable. Moreover, in
`patients with HIV infection, dose adjustments based on both ideal body weight and
`body surface did not affect this variability (23, 24). As a result of this variability,
`the pharmacokinetic properties of thalidomide in humans have not been well char-
`acterized, and this has confounded the definition of a dose-response effect against
`human cancer.
`
`DR. REDDY’S LABS., INC. EX. 1043 PAGE 3
`
`

`

`12 Dec 2001 8:13
`
`AR
`
`AR149-34.tex AR149-34.SGM
`
`LaTeX2e(2001/05/10)
`
`P1: GJC
`
`632
`
`RICHARDSON ¥ HIDESHIMA ¥ ANDERSON
`
`TABLE 2 Single dose pharmacokinetic parameters of thalidomide in humans (27)
`
`Mean apparent
`pharmacokinetic parameters
`
`Population
`
`Elderly patients with
`hormone-refractory
`prostate cancer
`Patients with HIV
`infection
`Healthy female volunteers
`Healthy male volunteers
`
`Dose
`
`200 mg
`
`800 mg
`300 mg
`
`200 mg
`200 mg
`
`tmax
`
`3.3*
`
`4.4*
`3.4
`
`5.8
`4.4
`
`t1=2 (L)
`
`Vd (L)
`
`6.5
`
`18.3
`5.7
`
`4.1
`8.7
`
`66.9
`
`165.8
`78.2
`
`53.0
`120.7
`
`Abbreviations: tmax, time to reach maximum concentrations; t1=2, elimination half-life; Vd, volume of distribution;
`HIV, human immunodeficiency virus.
`*Median value.
`
`Absorption
`When oral thalidomide at a dose of 100 mg/kg has been administered in animal
`studies, maximum serum concentrations were reached within 4 h (28). Absorption
`was apparently independent of the administered doses and slower than drug elim-
`ination. Recent studies in humans show a similar pattern; thalidomide at 200 mg
`per dose reaches peak concentration (tmax) in a mean of »4 h (23–25, 29).
`
`Distribution
`Animal studies have demonstrated a wide distribution of thalidomide throughout
`most tissues and organs (28). It is present in semen following oral administration
`in rabbits, but it is not known whether the drug is present in human semen (19, 30).
`Human pharmacokinetic studies to date also indicate that thalidomide has a large
`apparent volume of distribution (Vd) (24–26). Further, studies in elderly prostate
`cancer patients suggest variability in Vd, possibly due to alterations in absorption
`and plasma protein binding (24).
`
`Metabolism
`Thalidomide undergoes rapid and spontaneous nonenzymatic hydrolytic cleavage
`at physiologic pH to generate up to 50 metabolites, of which five are considered
`primary metabolites (8, 20, 22, 28, 31). Research efforts to better characterize the
`biologic properties of the specific metabolites have been complicated by their in-
`stability and rapid degradation under physiologic conditions (32). Whereas in vitro
`studies suggest thalidomide induces cytochrome P-450 isoenzymes in rats, recent
`evaluation of single- or multiple-dose pharmacokinetic parameters of oral thalido-
`mide at 200 mg daily in healthy human volunteers has indicated that thalidomide
`
`DR. REDDY’S LABS., INC. EX. 1043 PAGE 4
`
`

`

`12 Dec 2001 8:13
`
`AR
`
`AR149-34.tex AR149-34.SGM
`
`LaTeX2e(2001/05/10)
`
`P1: GJC
`
`THALIDOMIDE
`
`633
`
`does not inhibit or induce its own metabolism over a 21-day period in humans,
`and thus very little metabolism of thalidomide is thought to occur via the hepatic
`cytochrome P-450 system (26, 33, 34).
`
`Excretion
`Thalidomide appears to be rapidly excreted in urine as its metabolites, with the
`nonabsorbed portion of the drug excreted unchanged in feces. However, clearance
`is primarily nonrenal; mean terminal half-lives of the R and S isomers in healthy
`male human volunteers were measured at 4.6 and 4.8 h, respectively (23, 28, 29).
`In a study report of urinary excretion data for a single dose (200 mg daily), the
`elimination half-life was »8 h, with minimal drug excretion over a 24-h period
`(23). Both single and multiple dosing of thalidomide in older prostate cancer
`patients revealed a significantly longer half-life at a higher dose (1200 mg daily)
`than at a lower dose (200 mg daily) (24). Conversely, no effect of increased age
`on elimination half-life was identified in the age range of 55–80 years (24). Thus,
`the effects of renal or hepatic dysfunction on the clearance of thalidomide remain
`unclear, and additional studies are needed to better characterize age-related or
`physiologic effects on drug clearance.
`
`Drug Interactions
`The only drug that has been systematically evaluated for interaction with thalido-
`mide is oral hormonal contraceptives, which showed no significant interaction. An-
`imal studies suggest that thalidomide enhances the sedative effects of barbituates
`and alcohol as well as the catatonic effects of chlorpromazine and reserpine. Cen-
`tral nervous system stimulants (including methamphetamine and methylphenidate)
`appear to counteract the depressant effects of thalidomide (35).
`
`Potential Antitumor Effects
`D’Amato et al., while evaluating thalidomide’s mechanism of teratogenicity, found
`that it exhibited antiangiogenic properties (6, 7). They postulated that thalido-
`mide inhibited angiogenesis by interrupting processes induced by fl-FGF and/or
`VEGF (6, 7, 36). Further in vitro studies suggested that the antiangiogenic effect
`of thalidomide was due to specific metabolites and not the parent compound (37).
`Another important property of thalidomide is that it selectively inhibits TNF-fi
`production while leaving the patient’s immune system otherwise intact (38). This
`has led to its application in various disorders characterized by abnormal TNF-fi
`activity, including ENL, mycobacterium tuberculosis infection, graft-versus-host
`disease, rheumatoid arthritis, systemic lupus erythematosis, multiple sclerosis,
`Crohn’s disease, cancer- and HIV-related cachexia, diabetes mellitus, and endo-
`toxic shock.
`The exact mechanism of thalidomide-induced TNF-fi inhibition is unclear, but
`it does appear to differ from other TNF-fi inhibitors such as pentoxyfylline and
`
`DR. REDDY’S LABS., INC. EX. 1043 PAGE 5
`
`

`

`12 Dec 2001 8:13
`
`AR
`
`AR149-34.tex AR149-34.SGM
`
`LaTeX2e(2001/05/10)
`
`P1: GJC
`
`634
`
`RICHARDSON ¥ HIDESHIMA ¥ ANDERSON
`
`dexamethasone (39, 40). One mechanism postulated is that thalidomide inhibits
`TNF-fi synthesis by accelerating degradation of TNF-fi mRNA, resulting in a
`significant but incomplete suppression of TNF-fi protein production (39, 41). Of
`particular interest is the recent demonstration that thalidomide decreases the bind-
`ing activity of NF-•B which in turn controls activation of the TNF-fi gene (42).
`It has also been postulated that thalidomide’s effect on angiogenesis may be
`through TNF-fi inhibition, since TNF-fi has proangiogenic effects (6). However,
`the absence of a demonstrable TNF-fi effect in experimental models of angiogen-
`esis, coupled with the inability of strong TNF-fi inhibitors to directly influence
`angiogenesis, suggests that thalidomide’s antiangiogenic activity is not related to
`TNF-fi inhibition alone (6, 7).
`
`Immunomodulatory Effects
`Results of studies evaluating the effects of thalidomide on lymphocytes have been
`inconsistent (43–45). Emerging evidence suggests that thalidomide does not di-
`rectly suppress lymphocyte proliferation (14). However, differential effects on
`T cell stimulation, shifts in T cell responses, and inhibition of proliferation of
`already stimulated lymphocytes have been shown (17, 44, 46–48). Modification
`of surface adhesion molecule on leukocytes, inhibition of neutrophil chemotaxis,
`and effects of cytokines other than TNF-fi have been demonstrated, including
`the inhibition of interleukin (IL)-12 production, enhanced synthesis of IL-2, and
`inhibition of IL-6 (14, 16, 38, 49–51).
`
`Adverse Effects
`Common side effects reported during treatment with thalidomide are summarized
`in Table 3 (27). Sedation and constipation appear to be the most common adverse
`effects in cancer patients (9, 13, 36). The most serious adverse effect associated
`with thalidomide is peripheral neuropathy. There may also be an increased inci-
`dence of thromboembolic events, but this appears to be rare when the drug is used
`as a single agent. However, recent reports have shown thromboembolic complica-
`tions to be more frequent when the drug is combined with steroids and particularly
`
`TABLE 3 Clinical adverse events reported during thalidomide use (27)
`
`Neurologic
`
`Gastrointestinal
`
`Dermatologic
`
`Miscellaneous
`
`Sedation
`Dizziness
`Mood changes
`Headaches
`
`Constipation
`Nausea
`Increased appetite
`
`Exfoliative/erythrodermic
`Cutaneous reactions
`Brittle fingernails
`Pruritis
`
`Xerostomia
`Weight gain
`Edema of face/limbs
`Reduction in thyroid
`hormone secretion
`Hypotension
`
`DR. REDDY’S LABS., INC. EX. 1043 PAGE 6
`
`

`

`12 Dec 2001 8:13
`
`AR
`
`AR149-34.tex AR149-34.SGM
`
`LaTeX2e(2001/05/10)
`
`P1: GJC
`
`THALIDOMIDE
`
`635
`
`with anthracycline-based chemotherapy, causing early cessation of one study ex-
`ploring a combination of thalidomide with doxil and dexamethasone (52, 53). Pos-
`sible cardiovascular effects of thalidomide include bradycardia and hypotension.
`The risk of adverse cardiovascular events appears greater in older patients with
`coronary disease taking multiple blood-pressure-lowering medications along with
`thalidomide (27).
`It is well-known that thalidomide must not be used during pregnancy, and rec-
`ommended contraceptive methods must be used by both men and women of child-
`bearing potential. The System for Thalidomide Education and Prescribing Safety
`(STEPS), implemented to ensure the safe distribution of thalidomide, requires a
`patient’s compliance with contraception guidelines and mandatory surveillance
`procedures. In addition, all healthcare providers who plan to prescribe and/or dis-
`pense thalidomide must be registered with the program (4).
`Thalidomide-induced peripheral neuropathy commonly presents with numb-
`ness of the toes and feet, muscle cramps, weakness, signs of pyramidal tract in-
`volvement, and carpal tunnel syndrome (5, 19). Clinical improvement typically
`occurs upon prompt discontinuation of the drug, but long-standing sensory loss
`has been documented (54–57). Drug-related neuropathy is characterized as asym-
`metric, painful, peripheral paresthesia with sensory loss (5, 54–57). The risk of
`developing peripheral neuropathy during thalidomide treatment increases when
`high cumulative doses are administered and especially in the elderly (19).
`In cancer patients, thalidomide should be used with caution when there is a prior
`history of neuropathy and when used in combination with other agents known to
`have neurotoxic effects (27). However, whether the incidence of thalidomide-
`induced peripheral neuropathy is increased in cancer patients with a history of
`vinca alkaloid use remains unknown. Safety data from current phase I and II
`clinical trials of thalidomide in the treatment of solid tumors, multiple myeloma,
`and hematologic malignancies suggest that peripheral neuropathy occurs in 10%–
`30% of patients (10, 12, 13, 58).
`
`PRECLINICAL STUDIES OF THALIDOMIDE
`AND ITS ANALOGS IN MULTIPLE MYELOMA
`
`Although thalidomide was initially used to treat multiple myeloma because of
`its antiangiogenic effects, the mechanism of its antimyeloma activity appears to
`be more complex (Figure 2). Preclinical studies of thalidomide and its potent
`analogs (also known as immunomodulatory drugs, IMiDs) suggest that these drugs
`act against myeloma in several ways. First, there appears to be a direct effect on
`the myeloma cell and/or bone marrow stromal cell, which inhibits tumor growth
`and survival. Second, adhesion of myeloma cells to bone marrow stromal cells
`(BMSCs) triggers secretion of cytokines, which augment myeloma cell growth
`and survival (59–61) and confer drug resistance (62); importantly, thalidomide
`modulates adhesive interactions (14) and thereby may alter tumor cell growth,
`
`DR. REDDY’S LABS., INC. EX. 1043 PAGE 7
`
`

`

`12 Dec 2001 8:13
`
`AR
`
`AR149-34.tex AR149-34.SGM
`
`LaTeX2e(2001/05/10)
`
`P1: GJC
`
`636
`
`RICHARDSON ¥ HIDESHIMA ¥ ANDERSON
`
`Figure 2 Possible effect of thalidomide on myeloma cells’ and bone marrow stro-
`mal cells’ (BMSCs’) microenvironment in vivo. (A) Thalidomide directly inhibits
`myeloma cell growth. (B) Thalidomide inhibits myeloma cell adhesion to BMSCs.
`(C) Thalidomide blocks IL-6, TNF-fi, and IL-1fl secretion from BMSCs. (D) Thalido-
`mide blocks the ability of VEGF and fl-FGF to stimulate neovascularization of bone
`marrow. (E) Thalidomide induces IL-2 and INF-(cid:176) secretion from T cells.
`
`survival, and drug resistance. Third, cytokines secreted into the bone marrow mi-
`croenvironment by myeloma cells and/or BMSCs, such as IL-6, IL-1fl, IL-10,
`and TNF-fi may augment myeloma cell growth and survival (61, 63), and thalido-
`mide may alter their secretion and bioactivity (64). Fourth, thalidomide decreases
`the secretion of VEGF, IL-6 (65), and flFGF by myeloma and/or BMSCs.
`Since IL-6 is known to promote myeloma cell growth and survival, and VEGF
`has been demonstrated to induce myeloma cell migration (66), thalidomide may
`directly block tumor cell growth and migration, as well as inhibiting bone mar-
`row angiogenesis (67, 68). Recent studies have demonstrated the direct antitumor
`activity of thalidomide and IMiDs on myeloma cells (18). IMiDs induce growth
`inhibition of myeloma cells in a dose-dependent fashion, with an IC50 as low as
`0.1–1 „M. IMiDs are effective against myeloma cell lines resistant to conven-
`tional chemotherapeutic agents such as doxorubicin, melphalan, or mitoxantrone,
`as well as dexamethasone. Moreover, thalidomide and the IMiDs enhanced the
`antitumor activity of dexamethasone (Figure 3a); conversely, these effects were
`partially inhibited by IL-6 (Figure 3b), a potent myeloma growth and antiapop-
`totic factor. IMiDs also showed significant activity against drug-resistant patient
`myeloma cells (Figure 4). The observed clinical activity of thalidomide in patients
`
`DR. REDDY’S LABS., INC. EX. 1043 PAGE 8
`
`

`

`12 Dec 2001 8:13
`
`AR
`
`AR149-34.tex AR149-34.SGM
`
`LaTeX2e(2001/05/10)
`
`P1: GJC
`
`THALIDOMIDE
`
`637
`
`Figure 3 Effect of Dex and IL-6 on response of multiple myeloma cells to thalido-
`mide and the IMiDs. (a) MM.1S cells were cultured with 1.0 „M thalidomide,
`IMiD1, IMiD2, or IMiD3 in control media alone (⁄) or with 0.001 ( ), 0.01 ( ), and
`0.1 „M (¥) Dex. (b) MM.1S cells were cultured in control media alone and with 0.1
`and 1.0 „M thalidomide, IMiD1, IMiD2, or IMiD3 either in the presence (⁄) or ab-
`sence (¥) of IL-6 (50 ng/ml). In each case, 3H-TdR uptake was measured during the
`last 8 h of48-h cultures. Values represent the mean ( § SD) 3H-TdR (cpm) of triplicate
`cultures.
`
`with myeloma that is refractory to conventional therapies (69), coupled with our
`in vivo studies, suggests that thalidomide can overcome resistance to conventional
`treatments. In addition, our laboratory work suggests that dexamethasone can add
`to the antiproliferative effect of thalidomide and its analogs in vitro, suggesting
`potential utility of coupling these agents in novel therapeutics.
`
`DR. REDDY’S LABS., INC. EX. 1043 PAGE 9
`
`

`

`12 Dec 2001 8:13
`
`AR
`
`AR149-34.tex AR149-34.SGM
`
`LaTeX2e(2001/05/10)
`
`P1: GJC
`
`638
`
`RICHARDSON ¥ HIDESHIMA ¥ ANDERSON
`
`Figure 4 Effect of thalidomide and its analogs on DNA synthesis of patient multi-
`ple myeloma cells cultured with control media (⁄), or with 0.1 „M ( ), 1.0 „M ( ),
`10 „M ( ), and 100 „M (¥) thalidomide, IMiD1, IMiD2, or IMiD3. In each case,
`3H-TdR uptake was measured during the last 8 h of 48-h cultures. Values represent the
`mean (§ SD) 3H-TdR (cpm) of triplicate cultures.
`
`The mechanism of growth inhibition induced by thalidomide and its analogs is
`not totally understood. IMiDs, and to a lesser extent thalidomide, induce apopto-
`sis of myeloma MM.1S cells, as evidenced both by increased sub-G1 cells on PI
`staining and by increased annexin V–positive cells. In these cells, which character-
`istically have wild-type p53, exposure to these agents (and dexamethasone) down-
`regulates p21, thereby facilitating G1-to-S transition and enhanced susceptibility
`to apoptosis. Moreover, p27 is also upregulated by treatment with both thalido-
`mide and IMiDs (T. Hideshima, et al., unpublished data). This profound apop-
`totic effect may correlate with the fact that complete responses to thalidomide are
`occasionally observed. The mechanism of thalidomide/IMiDs-induced apoptosis
`
`DR. REDDY’S LABS., INC. EX. 1043 PAGE 10
`
`

`

`12 Dec 2001 8:13
`
`AR
`
`AR149-34.tex AR149-34.SGM
`
`LaTeX2e(2001/05/10)
`
`P1: GJC
`
`THALIDOMIDE
`
`639
`
`is also not entirely understood. However, our preliminary data indicate the par-
`ticipation of caspase-8 activation in thalidomide/IMiDs-induced apoptosis. IL-6
`overcomes the downregulation of p21 induced by these agents, consistent with the
`increase in DNA synthesis triggered by IL-6 even in the presence of these drugs. In
`contrast, in Hs Sultan cells and AS cells, derived from a multiple myeloma patient,
`(which are both wild-type and mutant p53), the IMiDs and thalidomide induce
`p21 and related G1 growth arrest (Figure 5), thereby conferring protection from
`apoptosis, as has been observed in other systems (70, 71). Prior studies showed that
`p21 was constitutively expressed in the majority of myeloma cells and inhibited
`proliferation in both a p53-dependent and -independent fashion (72).
`Previous reports that cells overexpressing p21 protein demonstrate chemore-
`sistance (73) further support the notion that a protective effect of G1 growth ar-
`rest is induced by thalidomide and IMiDs in Hs Sultan- and AS-patient-derived
`myeloma cells. Conversely, the frequent regrowth of progressive myeloma noted
`
`Figure 5 Effect of thalidomide and its analogs on p21 expression in multiple mye-
`loma (MM) cell lines and patient cells. (a) MM.1S cells were cultured with 10 „M
`of thalidomide, IMiD1, IMiD2, and IMiD3 for 48 h. MM.1S cells were also cultured
`with IL-6 (50 ng/ml) alone and with IMiD1, 10 „M Dex, and Dex plus IL-6. Cells
`were lysed, subjected to SDS PAGE, transferred to PVDF membrane, and blotted
`with anti-p21 Ab. The membrane was stripped and reprobed with anti-fi-tubulin Ab.
`(b) MM.1S, Hs Sultan, and patient MM cells were lysed and immunoprecipitated with
`wt-p53 and mt-p53 Ab, transferred to PVDF membrane, and blotted with anti-p53 Ab.
`
`DR. REDDY’S LABS., INC. EX. 1043 PAGE 11
`
`

`

`12 Dec 2001 8:13
`
`AR
`
`AR149-34.tex AR149-34.SGM
`
`LaTeX2e(2001/05/10)
`
`P1: GJC
`
`640
`
`RICHARDSON ¥ HIDESHIMA ¥ ANDERSON
`
`Figure 6 Effect of thalidomide and its analogs on growth and apoptotic signaling
`in myeloma cell lines, MM.1S and MM.1R cells. (a) MM.1S cells were cultured for
`48 h in media, with 50 „M of PD98059, and with 10 „M of IMiD1, IMiD2, or
`IMiD3. Cells were then triggered with 50 ng/ml of IL-6 for 10 min, lysed, transferred
`to PVDF membrane, and blotted with anti-phospho MAPK Ab. Blots were stripped
`and reprobed with anti-ERK2 Ab. (b) MM.1S and MM.1R cells were treated with
`thalidomide (100 „M), IMiD 1 (100 „M), or Dex (10 „M) and harvested at 12 h. Total
`cell lysates were subjected to immunoprecipitation with anti-RAFTK Ab and analyzed
`by immunoblotting with anti-P-Tyr Ab or anti-RAFTK Ab.
`
`clinically upon discontinuation of thalidomide treatment may correlate with re-
`lease of drug-related G1 growth arrest. We have previously shown that related
`adhesion focal tyrosine kinase (RAFTK, Figure 6) mediated dexamethasone-
`induced myeloma cell apoptosis. Thalidomide and IMiDs, like dexamethasone,
`induced tyrosine phosphorylation of RAFTK, suggesting a key role of RAFTK in
`thalidomide- and IMiDs-induced apoptosis. Activation of RAFTK was also found
`in dexamethasone-resistant, IMiDs-sensitive MM.1R myeloma cells.
`Recent studies have also demonstrated that myeloma cell lines and patient
`myeloma cells produce TNF-fi (63). TNF-fi in turn triggers IL-6 secretion from
`BMSCs in a dose-dependent fashion; importantly, TNF-fi is a more potent
`stimulus of IL-6 secretion than TGF-fl1 and VEGF (Figure 7). Remarkably, TNF-fi
`also induces NF-•B activation and expression of LFA-1, ICAM-1, VCAM-1,
`and MUC-1 on myeloma cell lines, as well as ICAM-1 and VCAM-1 on BMSCs.
`
`DR. REDDY’S LABS., INC. EX. 1043 PAGE 12
`
`

`

`12 Dec 2001 8:13
`
`AR
`
`AR149-34.tex AR149-34.SGM
`
`LaTeX2e(2001/05/10)
`
`P1: GJC
`
`THALIDOMIDE
`
`641
`
`Figure 7 TNF-fi secretion from IM-9 multiple myeloma (MM) cells stimulates IL-6
`secretion from bone marrow stromal cells (BMSCs). (a) IM-9 MM cells, BMSCs, and both
`together were cultured for 24 h (⁄) and 48 h (¥). (b) IM-9 myeloma cells, BMSCs, and
`both together were cultured for 48 h. (c) BMSCs were cultured for 48 h in the presence of
`media alone, VEGF (10 ng/ml), TNF-fi (10 ng/ml), or TGF-fl1 (10 ng/ml). (d ) Two multi-
`
`ple myeloma patient BMSCs (¥, x) were cultured for 24 h with TNF-fi (0.0001–10 ng/ml).
`TNF-fi (a) or IL-6 (b, c, d ) levels were measured in culture supernatants by ELISA. Values
`represent the mean (§ SD) of triplicate cultures.
`
`As a result, adherence of myeloma cells to BMSCs is significantly upregulated
`by TNF-fi–related induction of these adhesion molecules. Adherence of myeloma
`cells to BMSCs induces drug resistance of myeloma cells, induces IL-6 secre-
`tion in BMSCs, and activates p44/42 MAPK in myeloma cells, thereby promoting
`tumor cell growth. Because thalidomide and its analogs are potent inhibitors of
`TNF-fi production, they may not only act directly on myeloma cells but also
`act indirectly by inhibiting cytokine cross-talk in the myeloma/BMSC microen-
`vironment. In conclusion, TNF-fi induces an increase in proliferation, as well as
`MAPK/ERK activation, in myeloma cells, and it induces IL-6 secretion, as well
`as NF-•B activation, in BMSCs. TNF-fi induces adhesion molecules on myeloma
`
`DR. REDDY’S LABS., INC. EX. 1043 PAGE 13
`
`

`

`12 Dec 2001 8:13
`
`AR
`
`AR149-34.tex AR149-34.SGM
`
`LaTeX2e(2001/05/10)
`
`P1: GJC
`
`642
`
`RICHARDSON ¥ HIDESHIMA ¥ ANDERSON
`
`cells and BMSCs, increasing IL-6 secretion and the binding of myeloma cells to
`BMSCs; conversely, blockade of TNF-fi–induced NF-•B activation inhibits these
`sequelae. These studies confirm a central role for TNF-fi in the growth and sur-
`vival of myeloma cells in the bone marrow milieu and suggest the utility of novel
`therapeutics targeting TNF-fi in multiple myeloma.
`Keifer et al. recently reported that thalidomide inhibited NF-•B activation trig-
`gered by TNF-fi in Jurkat T cells (74). This inhibitory effect of thalidomide on
`NF-•B activation was mediated by suppression of I•B kinase (IKK) activity.
`Phosphorylation of I•Bfi, inhibitory molecule of NF-•B, by IKK is essential for
`degradation of I•Bfi. Since adherence of myeloma cells to BMSCs induces IL-6
`secretion from BMSCs via NF-•B activation (60), and TNF-fi induces adhesion
`molecules on both myeloma cells and BMSCs also via NF-•B activation, NF-•B
`may be a novel therapeutic target.
`A recent study demonstrated that thalidomide and IMiDs augment natural
`killer (NK) cell cytotoxicity in multiple myeloma (75). This report showed that
`thalidomide and IMiDs do not induce T cell proliferation alone but act as co-
`stimulators to trigger proliferation of anti-CD3-stimulated T cells from multiple
`myeloma patients, accompanied by an increase in interferon-(cid:176) and IL-12 secre-
`tion. Treatment of patient peripheral blood mononuclear cells with thalidomide
`or IMiDs triggered increased lysis of autologous myeloma cells. Furthermore, pa-

`C
`CD56
`cells in response to thalidomide/IMiDs
`tients showed an increase in CD3
`therapy. Lentzch et al. (76) demonstrated that thalidomide and IMiDs inhibited
`microvessel density in murine transplanted Hs Sultan cells, resulting in longer
`survival of IMiD-treated mice than of nontreated mice. These findings strongly
`support the antiangiogenic effect of thalidomide and IMiDs in vivo.
`In summary, extensive preclinical studies in multiple myeloma provide a com-
`pelling basis for the development and testing of thalidomide and the IMiDs in a
`new treatment paradigm to target both the tumor cell and the microenvironment,
`overcome classical drug resistance, and improve outcome in this presently incur-
`able disease.
`
`CLINICAL STUDIES IN MULTIPLE MYELOMA
`
`Despite recent advances in treatment, including transplantation, myeloma remains
`incurable and more effective therapies are clearly needed (77, 78). The develop-
`ment of resistance to chemotherapy and radiation, characteristic of myeloma, has
`spurred research in new biologically derived treatment strategies. The strategy of
`antiangiogenesis is based on observations that hematologic malignancies, such as
`multiple myeloma, are associated with intense neovascularization of bone marrow
`and thus may be angiogenesis-dependent (79, 80). However, although angiogene-
`sis appears well-established as a key component in the growth, progression, and
`metastatic spread of solid tumors (81), its role in hematologic malignancy remains
`to be defined, and only relatively recently has clinical investigation suggested its
`potential in this setting (67, 69, 80, 82–84).
`
`DR. REDDY’S LABS., INC. EX. 1043 PAGE 14
`
`

`

`12 Dec 2001 8:13
`
`AR
`
`AR149-34.tex AR149-34.SGM
`
`LaTeX2e(2001/05/10)
`
`P1: GJC
`
`THALIDOMIDE
`
`643
`
`In 1994, Vacca and colleagues reported a high correlation between the extent
`of bone marrow angiogenesis and the labeling index (LI) of marrow plasma cells
`and disease activity in patients with multiple myeloma (80). Subsequent stud-
`ies have confirmed extensive bone marrow vascularization in multiple myeloma
`(67, 69, 83, 84) and have associated poor prognosis with both elevated levels of an-
`giogenic cytokines, such as fl-FGF and VEGF, and increased bone marrow levels
`of mast cells, which secrete a variety of angiogenic factors (80, 83–85). More-
`over, recent reports have shown increased bone marrow angiogenesis in acute
`lymphoblastic leukemia (ALL) in children (79). Collectively, these findings have
`provided the rationale for the use of antiangiogenic drugs in the treatment of mul-
`tiple myeloma and other hematologic malignancies.
`Thalidomide therapy for advanced refractory myeloma began after an encour-
`aging initial experience in two patients at the University of Arkansas prompted
`a large phase II study, which assessed the efficacy and toxicity of single-agent
`thalidomide in relapsed and refractory multiple myeloma (69). The primary end-
`point of this trial was paraprotein response. Additional

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