throbber
Leukemia (2000) 14, 2–8
`2000 Macmillan Publishers Ltd All rights reserved 0887-6924/00 $15.00
`www.nature.com/leu
`
`REVIEW
`
`A hypothesis for the pathogenesis of myelodysplastic syndromes: implications for
`new therapies
`C Rosenfeld1 and A List2
`
`1Texas Oncology, PA, Dallas, TX; and 2University of Arizona Cancer Center, Tucson, AZ, USA
`
`To guide development of new clinical strategies, a review of
`recent investigations in the pathobiology of MDS was perfor-
`med. Articles were identified through a Medline search. Stud-
`ies, including reviews, are cited in the references. A multistep
`pathogenesis is proposed. (1) Targeted injury or mutation
`within hemopoietic stem cells may be followed by an immuno-
`logic response adversely affecting progenitor survival.
`(2)
`Accelerated proliferation and premature death of marrow cells
`is amplified by apoptogenic cytokines (TNF-a, Fas ligand). (3)
`Establishment of an abnormal clone associated with telomere
`shortening. (4) Disease progression associated with loss of
`tumor suppressor activity. Opportunities for therapeutic inter-
`ventions are possible at each step. Comparisons between the
`proposed pathogenesis of MDS and severe aplastic anemia
`(SAA) are also presented. Leukemia (2000) 14, 2–8.
`Keywords: myelodysplastic syndrome; acute nonlymphocytic leu-
`kemia; refractory anemia; preleukemia
`
`Introduction
`
`Three decades of investigations into the pathophysiology of
`the myelodysplastic syndromes (MDS) have confirmed the
`heterogenicity of MDS and highlighted the complexity in dis-
`ease biology.1 Recent advances in technology have yielded
`provocative observations. The objective of this review is to
`integrate clinical and laboratory findings into a working
`hypothesis for the development of idiopathic MDS, differen-
`tiate idiopathic MDS from SAA, and suggest new thera-
`peutic strategies.
`Interpretation of data from MDS studies remains problem-
`atic. Without a reliable disease marker, there can be questions
`regarding the accuracy of an MDS diagnosis.2 Additional
`problems arise when patients with disparate biologies are
`compared. For example, patients with idiopathic MDS and
`therapy-related MDS are sometimes included in the same data
`analyses. The same is true for FAB morphologic type and cyto-
`genetics. A potential source of ambiguity in laboratory studies
`derives from the mixture of normal and malignant progenitor
`cells which are known to coexist.3 The low number of poly-
`clonal progenitor cells in most cases suggests that such studies
`are valid. However, patients
`in chemotherapy-induced
`remission may re-establish polyclonal hemopoiesis.4,5
`Any hypothesis for the pathogenesis of MDS must support
`some long-standing clinical observations. Why are cytopenias
`present with hypercellular marrows? Why does MDS evolve
`more slowly than AML? Why is idiopathic MDS predomi-
`nantly a disease of the elderly? Why does MDS sometimes
`respond to therapies for SAA? Proposals for the pathogenesis
`of MDS have been suggested previously.6–9 A specific multi-
`step sequence for the development of adult-onset idiopathic
`
`Correspondence: C Rosenfeld, Texas Oncology, PA, 7777 Forest
`Lane, Building D 400, Dallas, Texas 75230, USA; Fax: 972–566–5819
`Received 28 June 1999; accepted 2 September 1999
`
`MDS based on cell culture, cytokine, molecular and clinical
`research is presented (Figure 1).
`
`Early events in evolution of MDS
`
`Three large (.150 index cases) epidemiologic studies suggest
`that radiation, smoking and occupational exposure to pesti-
`cides, organic chemicals and heavy metals are risk factors for
`the development of MDS.10–12 Prevention of MDS will require
`
`Figure 1
`A proposed multistep sequence for the development of
`idiopathic myelodysplastic syndrome. M-CSF, macrophage colony-
`stimulating factor; GM-CSF, granulocyte–macrophage colony-stimul-
`ating factor; G-CSF, granulocyte colony-stimulating factor; ATG, anti-
`thymocyte globulin; TNF, tumor necrosis factor.
`
`Dr. Reddy’s Laboratories, Inc. v. Celgene Corp.
`IPR2018-01504
`Exhibit 2019, Page 1
`
`(cid:211)
`

`

`further delineation of disease-associated toxins and possible
`polymorphisms in toxin metabolism that may predispose to
`a higher risk of MDS.13 Clearly, additional epidemiologic
`studies are needed.
`One proposal is that progenitor cells damaged by toxin
`exposure or spontaneous mutation evoke an immunologic
`response that further compromises progenitor cell growth and
`maturation. What
`is the evidence for the existence of an
`aberrant immunologic response when over 25 years of studies
`indicate a diminished immune state in MDS?14 Incubation of
`marrow cells with cyclosporine or removal of T cells enhance
`colony formation in some patients.15,16 Studies reported by
`Molldrem and colleagues16 at
`the NIH indicate that sup-
`pression of CFU-GM may be mediated by CD8+ cells directed
`against MHC class I restricted antigens. The anti-CFU-GM
`response does not appear to be mediated by MDS-derived
`immune cells since, most, but not all, studies indicate that T
`cells are not clonal in MDS.17–22 Clinical observations also
`support the notion of immune suppression of progenitor cell
`growth in MDS. Treatment with anti-thymocyte globulin or
`cyclosporine can improve
`cytopenias
`in select MDS
`patients.23–26 In contrast, attempts to augment an immunologic
`response with roquinimex or IL-2 have met with very limited
`suceess indicating that the diminished immune response may
`be the result rather than the cause of MDS.27,28
`Non-clonal lymphopoiesis provides indirect evidence for a
`lack of stem cell involvement in MDS. Using precursors sorted
`by flow cytometry and subsequent FISH to define clonal hem-
`opoiesis, primitive progenitors (CD34+, Thy1+)
`lacked the
`cytogenetic marker whereas more committed progenitors
`(CD34+, CD33+) display a clonal chromosome abnormality.29
`Conceivably, these non-clonal primitive stem cells could be
`utilized as a stem cell graft for autologous transplantation.
`The growth and differentiation of the progeny of clonal pro-
`genitors is further compromised by an accelerated rate of
`apoptotic cell death. In cell culture, cytokines such as TNF-a
`and IFN-gcan suppress the growth of hemopoietic progenitors
`and induce Fas expression on CD34 cells.30–32 What is the
`evidence for a functional role of apoptogenic cytokines in
`MDS? Elevated serum levels of TNF-a in patients with MDS
`is well documented (see Table 1).33–35 Increased TNF-a pro-
`duction by blood mononuclear cells in one study was restric-
`ted to patients with RA and RARS, but not RAEB or RAEBt.36
`
`Table 1
`
`Cytokine levels in MDS compared to normal controls
`
`3
`
`Pathogenesis of MDS
`C Rosenfeld and A List
`
`Furthermore, overexpression of TNF-a mRNA from marrow
`was detected in most cases of MDS, but not in normal controls
`or AML patients.37 One probable source of TNF-a overpro-
`duction is marrow macrophages which are increased in
`MDS.38,39 The increased density of marrow macrophages may
`occur in response to elevated serum levels of M-CSF.40 Point
`mutations in c-fms, which encodes the M-CSF receptor, may
`also promote macrophage development in some cases.41 The
`physiological significance of TNF-a in MDS is supported by
`several lines of investigation: (1) enhanced in vitro formation
`of CFU-GM by antibody neutralization of TNF-a in MDS but
`no effect on AML CFU;42 (2)
`inverse correlation between
`serum TNF-a concentration and hemoglobin in one study;34
`(3) inverse correlation between clinical response to erythro-
`poietin and TNF-a levels;35 (4) inverse correlation between a
`platelet response to IL-3 therapy and TNF-a serum levels;13
`(5) positive correlation between TNF-a producing cells in the
`marrow and apoptosis;44 and (6) correlation between plasma
`TNF-a concentration with nucleotide oxidation in marrow
`MDS CD34+ cells.45
`This model suggests that secretion of TNF-a or other pro-
`apoptotic cytokines plays a pivotal role in the ineffective hem-
`atopoiesis of MDS, but the relationship to disease progression
`remains ill defined. This implies that strategies which effec-
`tively neutralize TNF may inprove hematopoiesis. Pentoxifyl-
`line at micromolar concentrations suppresses TNF-a mRNA
`transcription. Combination therapy with pentoxifylline + cip-
`rofloxacin yielded no hematologic benefit in one study but a
`triple drug regimen of pentoxifylline + ciprofloxacin +
`dexamethasone produced hemopoietic responses in 35%
`(18/51) of patients and 28% (5/18) of responders demonstrated
`a cytogenetic response.46,47 An alternative approach is to neu-
`tralize circulating TNF-a by administration of soluble TNF
`receptors. In vitro, incubation of MDS marrow with TNFR:Fc
`enhanced CFU-GM formation.42 Strategies which reduce the
`impact of multiple soluble mediators of progenitor cell
`apoptosis or increase the threshold for apoptosis induction
`may be more effective than single agent therapy. For instance,
`interruption of progenitor cell apoptosis could be attempted
`with soluble TNF receptor
`(to inhibit
`the initiation of
`apoptosis) plus amifostine (to raise the threshold for
`apoptosis). Another possibility for combined therapy is simul-
`taneous inactivation of more than one inducer of apoptosis.
`
`Colony-stimulating factors
`
`Pro-apoptotic factors
`
`Marrow cells
`
`Serum
`
`Blood cells
`
`Marrow cells
`
`Blood cells
`#79
`
`"36*
`
`Serum
`"34
`"76, U34
`"40
`"34, U76
`"34
`#75,83
`"85*
`
`G-CSF
`GM-CSF
`M-CSF
`IL-3
`IL-6
`SCF
`BPA
`FLT3 ligand
`"37,39, N80,82, U51
`TNF-a
`"42, "59‡
`Fas ligand
`"39,51
`IL-1b
`"39, N51
`TGF-b
`"37, U49
`IFN-g
`", increased; #, decreased; N, not different from normal controls; U, undetectable; *increased predominantly in patients with refractory
`anemia; ‡levels higher in RAEB/RAEBt than in RA/RARS.
`
`U51
`#80,82, U51, N39
`
`"51, N80
`#84
`
`"33–35, "42*
`"35
`
`"36*
`"36*
`
`Leukemia
`
`Dr. Reddy’s Laboratories, Inc. v. Celgene Corp.
`IPR2018-01504
`Exhibit 2019, Page 2
`
`

`

`Pathogenesis of MDS
`C Rosenfeld and A List
`
`One potential approach includes simultaneous blockade of
`the activity of Fas ligand and TNF-a.48
`IFN-g or IL-1b are apoptogenic cytokines that could con-
`tribute to ineffective hematopoiesis in MDS. In two series,
`IFN-g gene overexpression was detected in only 5/12 and
`0/11 MDS cases.37,49 Increased production of IL-1b by blood
`and marrow cells has been reported.36,50,51 Increased pro-
`duction of IL-1b by cultured marrow mononuclear cells was
`detected in 13/32 MDS patients.50 Patients with RA tended to
`have the highest IL-1b production.36,50 IL-1blevels have been
`correlated with the extent of apoptosis, but not proliferation.50
`Deficient production of the IL-1b receptor antagonist by MDS
`stromal cells may give rise to unopposed apoptotic activity
`from IL-1b.52 These studies suggest that IFN-g and IL-1b do
`not contribute to apoptosis in the majority of patients with
`MDS.
`Several investigators have reported increased marrow cell
`apoptosis in MDS and have implicated the potential role of
`the Fas/Fas ligand system.53–58 Increased Fas expression was
`detected on marrow CD34 cells from MDS patients.42,55,56
`Lack of correlation between Fas expression and apoptosis sug-
`gests that multiple mediators of cell death are operational.55
`Gupta and coinvestigators59 have examined Fas
`ligand
`expression in marrows of MDS patients. The mean percentage
`of Fas ligand expressing cell was higher in MDS (17%) than
`from normal controls (6%). In contrast to normals where Fas
`ligand was detected mostly in lymphocytes, Fas ligand was
`expressed in erythroblasts, myeloblasts, megakaryocytes, mat-
`uring myeloid cells and dysplastic cells. In another study, Fas
`ligand expression in MDS patients was localized to marrow
`macrophages.60 Further
`investigations revealed more Fas
`ligand positive marrow cells in RA/RARS (9%)
`than in
`RAEB/RAEBt (20%).59 However, this appears to be inconsist-
`ent with the finding that the extent of marrow cell apoptosis
`inversely correlates with clinical stage of MDS. A higher
`degree of apoptosis was seen in early FAB classes for both
`CD34+ cells or marrow aspirates in most studies.53–56 Further-
`more, as MDS clinically progresses, apoptotic signals decrease
`(Fas antigen, c-Myc oncoprotein) whereas anti-apoptotic sig-
`nals increase (bcl-2 oncoprotein).54,55,61 Fas-associated phos-
`phatase-1 (fap-1) is a negative regulator of fas. Recent studies
`have shown that fap-1 expression is reduced in MDS marrow
`cells compared to marrow cells from either normals or AML
`than has progressed from MDS.62 Several investigators have
`suggested that the clinical sequelae of apoptosis is cytopenia.
`Since MDS is usually detected by cytopenia and apoptotic
`activity is most pronounced in the early phases of MDS, it is
`possible that apoptosis precedes the clinical recognition of
`MDS.
`Amifostine is a phosphorylated aminothiol with dual bio-
`logic activities including free radical scavenging, by addition
`of reducing equivalents, and inhibition of TNF-a and other
`inflammatory cytokine elaboration.63 In this way, amifostine
`may protect against TNF-induced apoptosis in MDS.43 In one
`trial, amifostine responses were noted in 83% (15/18)
`patients.64 In another study with 12 patients, none satisfied
`the criteria for a partial or complete response.65 Theoretically,
`amifostine activity should be more pronounced in early, rather
`than, late MDS. However, the potential application of this
`agent
`in MDS awaits further
`investigation.
`Inhibition of
`apoptosis by agents that decrease the c-Myc/Bcl-2 ratio could
`also be attempted. There are candidate agents to decrease
`c-Myc (Vitamin D3, agents
`that potentiate intracellular
`cAMPcontent and antisence oligonucleotide).66–68
`Extensive cell culture studies have been performed to inves-
`
`tigate the cause(s) of cytopenia in established MDS. Numerous
`studies indicate deficient growth of myeloid, erythroid and
`megakaryocytic colonies.69 Similar to AML, blast cell colonies
`(CFU-L) can be detected in MDS.70 One cause for cytopenias
`may be related to diminished capacity for differentiation.71
`Therapeutic trials of differentiating agents have not demon-
`strated consistent efficacy in ameliorating cytopenia.72–74
`Interpretation of the results from investigations of colony-
`stimulating factor levels in MDS patients is difficult to discern.
`Results appear inconsistent (Table 1) and CSF levels may be
`altered by clinical events (ie infections). Furthermore, in most
`studies, serum CSF levels did not correlate with clinical para-
`meters.34,75,76 Given these limitations, the decreased cellular
`production of G-CSF, GM-CSF and burst-promoting activity
`(BPA) plus low serum levels of SCF suggest that low CSF levels
`may be related to cytopenias and provide a basis for CSF ther-
`apy of MDS. Diminished elaboration of CSF may arise from
`apoptosis and functional abnormalities of stromal cells.44,77,78
`Studies indicate decreased production of GM-CSF and G-CSF
`by MDS monocytes.79,80 The use of colony-stimulating factors
`for the treatment of MDS is beyond the scope of this paper.81
`
`MDS disease progression
`
`The next proposed step is reduction of telomere length. Tel-
`omeres are located at the ends of eukaryotic chromosomes,
`and function to stabilize chromosomes. Telomere length is
`progressively reduced by cell divisions. Accelerated apoptosis
`and proliferation may lead to the reduction in telomere length
`observed in MDS.53,86,87 Reduction in telomere length is prob-
`ably not directly responsible for disease progression since only
`42% of patients with RA have telomere length reduction.87
`Instead, telomere shortening may give rise to genomic insta-
`bility that leads to cytogenetic evolution and disease pro-
`gression. This assertion is supported by the finding that telo-
`mere
`shortening
`is
`associated with advanced MDS,
`cytogenetic abnormalities, percentage of marrow blasts, leu-
`kemic transformation and poor prognosis.86,87 Since telomer-
`ase activity (a DNA polymerase that can synthesize the telo-
`meric sequence) is normal to low in most patients, telomere
`length may be a better reflection of the pathophysiology in
`MDS.86 Progenitor cells with shortened telomeres may be
`more susceptible to elimination by telomerase inhibitors.
`Telomerase inhibitors are currently in development.88
`Progression to advanced MDS and AML has been linked to
`inactivation of the tumor suppressor genes, p15INK4b and to a
`lesser extent, p53, and thereby contribute to clonal expansion
`(see Figure 1). p53 is a tumor suppressor gene which serves
`as a major control of the G1 checkpoint.89 Studies indicate that
`mutations of p53 occur in less than 20% of MDS cases.90–93 At
`the molecular level, p53 mutations in MDS are usually point
`or missense mutations of one allele, associated in some cases
`with 17 p deletion of the alternante allele.94 Evidence that sug-
`gests mutation of p53 is related to progression of MDS
`includes the limitation to advanced MDS (RAEB, RAEBt),
`association with complex cytogenetic abnormalities, and risk
`of secondary leukemia.89–91 In view of the low frequency of
`mutations, introduction of wild-type p53 into MDS cells by
`gene therapy offers limited therapeutic potential.95 Bishop and
`coinvestigators96 investigated whether a rebound in p53 after
`brief inactivation of p53 RNA by antisense oligonucleotide
`could inhibit clonal proliferation.
`In a phase I
`trial
`that
`included 10 high risk MDS patients (three RAEB, seven RAEB-
`t), there was one transient response detected after intravenous
`infusion OL(1)p 53.
`
`4
`
`Leukemia
`
`Dr. Reddy’s Laboratories, Inc. v. Celgene Corp.
`IPR2018-01504
`Exhibit 2019, Page 3
`
`

`

`As noted earlier, Fas expression decreases as the blast per-
`centage increases.55 The inhibitory effect of TGFb on leu-
`kemic colony formation in early MDS is diminished after
`transformation to AML.97 Transcription of p15INK4b, a cyclin-
`dependent kinase inhibitor represents one mechanism by
`which TGFb exerts its inhibitory effect. Hypermethylation of
`the p 15INK4b gene occurs in 38–50% of MDS patients and
`may contribute to loss of proliferative regulation.98,99 Evidence
`implicating a causal role for inactivation of p 15INK4b with dis-
`ease progression is provided by the increased frequency of
`hypermethylation observed in advanced MDS and secondary
`AML compared to early MDS. Agents which promote hypo-
`methylation of DNA may impact the evolution of the leukemic
`clone by derepression of p 15 transcription.100 5-Aza-29-
`deoxycytidine and 5-azacytidine promote DNA hypomethyl-
`ation by inhibition of DNA methyltransferase.101 However,
`most of the activity of 5-azacytidine, a drug demonstrated to
`have clinical efficacy, is through mechanisms other than DNA
`demethylation.101,102
`Incubation of cell
`lines harboring
`methylation silenced p 15 with 5-Aza-29-deoxycytidine leads
`to re-expression of p 15.103 In one study, a 72 h infusion of
`5-Aza-29-deoxycytidine to 29 high risk MDS patients yielded
`a 29% complete response rate and 24% partial responses.104
`Prolonged myelosuppression was the sole cause for a 17%
`drug-related mortality. Using a lower dose of 5-Aza-29-deoxy-
`cytidine in 61 patients produced similar clinical effects with-
`out marked myelosuppression.105 Additional studies with 5-
`Aza-29-deoxycytidine should be performed.
`
`Clinical relevance of model
`
`How does this model account for MDS being a disease of
`the elderly? Accumulated environmental exposures provide a
`cumulative probability of mutational events that
`increases
`with time. Age-related telomere shortening fosters heightened
`susceptibility to genomic instability that can lead to emerg-
`ence of clonal disease. A similar explanation can account for
`the differences between MDS and SAA. The younger age of
`SAA patients may reflect exposure to a progenitor cell toxin
`or a genetic event in a patient not susceptible to genomic
`instability from age-related telomere loss. This proposal is sup-
`ported by the finding of shorter telomere lengths in MDS than
`SAA.106 Another difference between MDS and SAA may be
`the milieu of hemopoietic inhibitory cytokines that drive
`apoptosis. In SAA, IFN-g gene overexpression was detectable
`in most patients.49,107 As described earlier, IFN-g mRNA was
`usually not detected in MDS patients, but TNF-a was over-
`expressed in 11/14 cases.37,49 These data suggest a tendency
`IFN-g-mediated apoptosis in SAA and TNF-a-driven
`for
`apoptosis in MDS. Some features of MDS and SAA are shown
`in Table 2. We propose that MDS and SAA may have a similar
`
`Table 2
`
`Pathophysiologic factors in MDS and SAA
`
`" Hemopoietic inhibitors
`#Progenitor cells
`Apoptosis of marrow cells
`Telomere shortening
`Clonal hemopoiesis
`Production of G-CSF and GM-
`CSF
`
`MDSa
`
`Yes
`Yes
`Yes
`Yesb
`Yes
`Often #
`
`SAA
`
`Yes36,49,108
`Yes109
`Yes110,111
`Yes106
`Rare112
`Normal or "113
`
`aSee text for details.
`bMore pronounced telomere shortening in MDS than SAA (see text).
`
`Pathogenesis of MDS
`C Rosenfeld and A List
`
`pathophysiology but the disease expression is dependent on
`patient age.
`There are a few other points suggested by the model. Mar-
`row failure has a multifactorial etiology including decreased
`marrow production of colony-stimulating factors, increased
`elaboration of hemopoietic inhibitors and accelerated
`apoptosis of progenitor cells. As proposed previously, cyto-
`penia with a hypercellular marrow can be attributed to simul-
`taneous progenitor cell apoptosis, increased proliferation with
`a differentiation block.44,53,71
`The sequential development of MDS does not explain all
`observations. This proposal provides a basis for designing new
`studies for what remains an enigmatic disease.
`
`References
`
`1 Greenberg PL, Nichols WC, Schrier SL. Granulopoiesis in acute
`myeloid leukemia and preleukemia. New Engl J Med 1971; 22:
`1225–1232.
`2 Rosati S, Anastasi J, Vardiman J. Recurring diagnostic problems in
`the pathology of the myelodysplastic syndromes. Semin Hematol
`1996; 33: 111–126.
`3 Asano H, Ohashi H, Ichihara M, Kinoshita T, Murate T, Kobayashi
`M, Saito H, Hotta T. Evidence for nonclonal hematopoietic pro-
`genitor cell populations in bone marrow of patients with myelo-
`dysplastic syndromes. Blood 1994; 84: 588–594.
`4 Delforge M, Demuynck H, Vandenberghe P, Verhoef G, Zache´e
`P, Van Duppen V, Marijnen P, Van den Berghe H, Boogaerts M.
`Polyclonal primitive hematopoietic progenitors can be detected in
`mobilized peripheral blood from patients with high-risk myelo-
`dysplastic syndromes. Blood 1995; 86: 3660–3667.
`5 Delforge M, Demuynck H, Verhoef G, Vandenberghe P, Zache´e
`P, Maertens J, Duppen VV, Boogaerts MA. Patients with high risk
`myelodysplastic syndrome can have polyclonal or clonal haemo-
`poiesis in complete haematological remission. Br J Haematol
`1998; 102: 486–494.
`6 List AF, Jacobs A. Pathogenesis and biology of myelodysplasia.
`Semin Oncol 1992; 29: 14–24.
`7 Sanz GF, Sanz MA, Vallespi T. Etiopathogeny, prognosis and ther-
`apy of myelodysplastic syndromes. Hematol Cell Ther 1997; 39:
`277–294.
`8 Gallagher A, Darley RL, Padua R. The molecular basis of myelo-
`dysplastic syndromes. Haematologica 1997; 82: 191–204.
`9 Aul C, Bowen DT, Yoshida Y. Pathogenesis, etiology and epidemi-
`ology of myelodysplastic syndromes. Haematologica 1998; 83:
`71–86.
`10 West RR, Stafford DA, Farrow A, Jacobs A. Occupational and
`environmental exposures and myelodysplasia: a case–control
`study. Leukemia Res 1995; 19: 127–139.
`11 Nisse C, Grandbastien B, Brizard A, Hebbar M, Haguenoer JM,
`Fenaux P. Myelodysplastic syndromes (MDS) and exposure to
`occupational or environmental hazards: a case–control study.
`Blood 1997; 90 (Suppl. 1): 518a (Abstr.).
`12 Rigolin GM, Cuneo A, Roberti MG, Bardi A, Bigoni R, Piva N,
`Minotto C, Agostini P, Angeli CD, Senno LD. Exposure to myelo-
`toxic agents and myelodysplasia: case–control study and corre-
`lation with clinobiologic findings. Br J Haematol 1998; 103:
`189–197.
`13 Larson LA, Wang Y, Banerjee M, Wiemels J, Hartford C, Le Beau
`MM, Smith MT. Prevalence of the inactivating609C-.T polymor-
`phism in the NAD(P)H: quinone oxidoreductase (NQO1) gene in
`patients with primary and therapy-related myeloid leukemia.
`Blood 1999; 94: 803–807.
`14 Takaku S, Takaku F. Natural killer cell activity and preleukaemia.
`Lancet 1981; 2: 1178.
`15 List AF, Glinsmann-Gibson B, Spier C, Taetle R. In vitro and in
`vivo response to cyclosporin-A in myelodysplastic syndromes
`(MDS):
`identification of a hypocellular subset
`responsive to
`immune suppression. Blood 1992; 80 (Suppl. 1): 28a (Abstr.).
`16 Molldrem JJ, Jiang YZ, Stetler-Stevenson M, Mavroudis D, Hensel
`N, Barrett AJ. Hematological response of patients with myelo-
`dysplastic syndrome to antithymocyte globulin is associated with
`
`5
`
`Leukemia
`
`Dr. Reddy’s Laboratories, Inc. v. Celgene Corp.
`IPR2018-01504
`Exhibit 2019, Page 4
`
`

`

`Pathogenesis of MDS
`C Rosenfeld and A List
`
`a loss of lymphocyte-mediated inhibition of CFU-GM and alter-
`ations in T-cell receptor Vb profiles. Br J Haematol 1998; 102:
`1314–1322.
`17 Van Kamp H, Fibbe WE, Jansen RPM, van der Keur M, de Graaff
`E, Willemze R, Landegent JE. Clonal involvement of granulocytes
`and monocytes, but not of T and B lymphocytes and natural killer
`cells in patients with myelodysplasia: analysis by X-linked restric-
`tion fragment length polymorphisms and polymerase chain reac-
`the phosphoglycerate kinase gene. Blood 1992; 80:
`tion of
`1774–1780.
`18 Hamblin TJ. Immunological abnormalities in myelodysplastic syn-
`dromes. Semin Hematol 1996; 33: 150–162.
`19 Tefferi A, Thibodeau SN, Solberg LA. Clonal studies in the myelo-
`dysplastic syndrome using X-linked restriction fragment length
`polymorphisms. Blood 1990; 75: 1770–1773.
`20 Kibbelaar RE, van Kamp H, Dreef EJ, de Groot-Swings G, Kluin-
`Nelemans JC, Beverstock GC, Fibbe WE, Kluin M. Combined
`immunophenotyping and DNA in situ hybridization to study lin-
`eage involvement in patients with myelodysplastic syndromes.
`Blood 1992; 79: 1823–1828.
`21 Anastasi J, Feng J, Le Beau M, Larson RA, Rowley JD, Vardiman
`JW. Cytogenetic clonality in myelodysplastic syndromes studied
`with fluorescence in situ hybridization:
`lineage, response to
`growth factor therapy, and clone expansion. Blood 1993; 81:
`1580–1585.
`22 Kroef MJPL, Fibbe WE, Mout R, Jansen RPM, Haak HL, Wessels
`JW, Van Kamp H, Willemze R, Landegent JE. Myeloid but not
`lymphoid cells carry the 5 q deletion: polymerase chain reaction
`analysis of loss of heterozygosity using mini-repeat sequences on
`highly purified cell fractions. Blood 1993; 81: 1849–1854.
`23 Sulecki M, Shadduck RK, Zeigler Z. Anti-thymocyte globulin for
`hypoplastic myelodysplastic syndrome. Blood 1988; 72 (Suppl. 1):
`229a (Abstr.).
`24 Biesma DH, van den Tweel JG, Verdonck LF. Immunosuppressive
`therapy for hypoplastic myelodysplastic syndrome. Cancer 1997;
`79: 1548–1551.
`25 Molldrem J, Caples M, Mavroudis D, Plante M, Young NS, Barrett
`AJ. Antithymocyte globulin for patients with myelodysplastic syn-
`drome. Br J Haematol 1997; 99: 699–705.
`26 Jona´s¯ova´ A, Neuwirtova´ R, C¯ erma´k J, Vozobulova´ V, Mocikova´
`K, S˘is¯kova´ M, Hochova´ I. Cyclosporin A therapy in hypoplastic
`MDS patients and certain refractory anaemias without hypoplastic
`bone marrow. Br J Haematol 1998; 100: 304–309.
`27 Rosenfeld CS, Zeigler ZR, Shadduck RK, Nilsson B. Phase II study
`of roquinimex in myelodysplastic syndrome. Am J Clin Oncol
`1997; 20: 189–192.
`28 Nand S, Stock W, Stiff P, Sosman J, Martone B, Radvany R. A
`phase II trial of interleukin-2 in myelodysplastic syndromes. Br J
`Haematol 1998; 101: 205–207.
`29 Saitoh K, Miura I, Takahashi N, Miura AB. Fluorescence in situ
`hybridization of progenitor cells obtained by fluorescence-acti-
`vated cell sorting for the detection of cells affected by chromo-
`some abnormality trisomy 8 in patients with myelodysplastic syn-
`dromes. Blood 1998; 92: 2886–2892.
`30 Selleri C, Sato T, Anderson S, Young NS, Maciejewski JP. Inter-
`feron-gamma and tumor necrosis factor-alpha suppress both early
`and late stages of hematopoiesis and induce programmed cell
`death. J Cell Physiol 1995; 165: 538–546.
`31 Nagafuji K, Shibuya T, Harada M, Mizuno S, Takenaka K, Miya-
`moto T, Okamura T, Gondo H, Niho Y. Functional expression of
`Fas antigen (CD95) on hematopoietic progenitor cells. Blood
`1995; 86: 883–889.
`32 Maciejewski
`J, Selleri C, Anderson S, Young NS. Fas antigen
`expression on CD34+ human marrow cells is induced by inter-
`feron g and tumor necrosis factor a and potentiates cytokine-
`mediated hematopoietic suppression in vitro. Blood 1995; 85:
`3183–3190.
`33 Zoumbos N, Symeonidis A, Kourakli A, Katevas P, Matsouka P,
`Perraki M, Georgoulias V. Increased levels of soluble interleukin-
`2 receptors and tumor necrosis factor in serum of patients with
`myelodysplastic syndromes. Blood 1991; 77: 413–414.
`34 Verhoef GEG, De Schouwer P, Ceuppens JL, Van Damme J, Goos-
`sens W, Boogaerts MA. Measurement of serum cytokine levels in
`patients with myelodysplastic syndromes. Leukemia 1992; 12:
`1268–1272.
`
`35 Stasi R, Brunetti M, Bussa S, Conforti M, Martin LS, Presa ML,
`Bianchi M, Parma A, Pagano A. Serum levels of tumour necrosis
`factor-apredict response to recombinant human erythropoietin in
`patients with myelodysplastic syndrome. Clin Lab Haematol 1997;
`19: 197–201.
`36 Koike M, Ishiyama T, Tomoyasu S, Tsuruoka N. Spontaneous cyto-
`kine overproduction by peripheral blood mononuclear cells from
`patients with myelodysplastic syndromes and aplastic anemia.
`Leukemia Res 1995; 19: 639–644.
`37 Kitagawa M, Saito I, Kuwata T, Yoshida S, Yamaguchi S, Takahaski
`M, Tanizawa T, Kamiyama R, Hirokawa K. Overexpression of
`tumor necrosis factor (TNF)-aand interferon (IFN)-gby bone mar-
`row cells from patients with myelodysplastic syndromes. Leuke-
`mia 1997; 11: 2049–2054.
`38 Kitagawa M, Kamiyama R, Kasuga T. Increase in number of bone
`marrow macrophages in patients with myelodysplastic syndromes.
`Eur J Haematol 1993; 51: 56–58.
`39 Shetty V, Mundle S, Alvi S, Showel M, Broady-Robinson L, Dar
`S, Borok R, Showel J, Gregory S, Rifkin S, Gezer S, Parcharidou
`A, Venugopal P, Shah R, Hernandez B, Klein M, Alston D, Robin
`E, Dominquez C, Raza A. Measurement of apoptosis, proliferation
`and three cytokines in 46 patients with myelodysplastic syn-
`dromes. Leukemia Res 1996; 20: 891–900.
`40 Janowska-Wieczorek A, Belch AR, Jacobs A, Bowen D, Padua RA,
`Paietta E, Stanley ER. Increased circulating colony-stimulating fac-
`tor-1 in patients with preleukemia, leukemia, and lymphoid malig-
`nancies. Blood 1991; 77: 1796–1803.
`41 Tobal K, Pagliuca A, Bhatt B, Bailey N, Layton DM, Mufti GJ.
`Mutation of the human FMS gene (M-CSF receptor) in myelodys-
`plastic syndromes and acute myeloid leukemia. Leukemia 1990;
`4: 486–489.
`42 Gersuk GM, Beckham C, Loken MR, Kiener P, Anderson JE, Far-
`rand A, Trout AB, Ledbetter JA, Deeg HJ. A role for tumour
`necrosis factor-alpha, Fas, and Fas-Ligand in marrow failure asso-
`ciated with myelodysplastic syndrome. Br J Haematol 1998; 103:
`176–188.
`43 Ganser A, Ottmann OG, Seipelt G, Lindemann A, Hess U, Geissler
`G, Maurer A, Frisch J, Schulz G, Mertelsmann R, Hoelzer D. Effect
`of long-term treatment with recombinant human interleukin-3 in
`patients with myelodysplastic syndromes. Leukemia 1993; 7:
`696–701.
`44 Raza A, Mundle S, Shetty V, Alvi S, Chopra H, Span L, Parchari-
`dou A, Dar S, Venugopal P, Borok R, Gezer S, Showel J, Loew J,
`Robin E, Rifkin S, Alston D, Hernandez B, Shah R, Kaizer H, Gre-
`gory S. Novel insights into the biology of myelodysplastic syn-
`dromes: excessive apoptosis and the role of cytokines. Int J Hema-
`tol 1996; 63: 265–278.
`45 Peddie CM, Wolf CR, McLellan LI, Collins AR, Bowen DT. Oxidat-
`ive DNA damage in CD34+ myelodysplastic cells is associated
`with intracellular redox changes and elevated plasma tumour
`necrosis factor-a concentration. Br J Haematol 1997; 99: 625–
`631.
`46 Nemunaitis J, Rosenfeld C, Getty L, Boegel F, Meyer W, Jennings
`LW, Zeigler Z, Shadduck R. Pentoxifylline and ciprofloxacin in
`patients with myelodysplastic syndrome. Am J Clin Oncol 1995;
`18: 189–193.
`47 Raza A, Gezer S, Venugopal P, Kaizer H, Hines C, Thomas R, Alvi
`S, Mundle S, Shetty V, Borok R, Lowe J, Reza S, Robin EL, Rifkin
`SD, Alston D, Hernandez B, Shah R, Hsu WT, Dar S, Gregory SA.
`Hematopoietic and cytogenetic responses to novel anti-cytokine
`therapy in myelodysplastic syndromes (MDS). Proc ASCO 1997;
`16: 7a (Abstr.).
`48 Hattori K, Hirano T, Miyajima H, Yamakawa N, Tateno M, Oshimi
`K, Kayagaki N, Yagita H, Okumura K. Differential effects of anti-
`Fas ligand and anti-tumor necrosis factor a antibodies on acute
`graft-versus-host disease pathologies. Blood 1998; 91: 4051–
`4055.
`49 Nistico` A, Young NS. Gamma-interferon gene expression in the
`bone marrow of patients with aplastic anemia. Ann Intern Med
`1994; 120: 463–469.
`50 Mundle SD, Venugopal P, Cartlidge JD, Pandav DV, Broday-Rob-
`inson L, Gezer S, Robin EL, Rifkin SR, Klein M, Alston DE, Hernan-
`dez BM, Rosi D, Alvi S, Shetty VT, Gregory SA, Raza A. Indication
`of an involvement of interleukin-1b converting enzyme-like pro-
`tease in intramedullary apoptotic cell death

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