throbber
| MyetopysPLastic SYNDROMES: UNDERSTANDING THE CURRENT TREATMENT LANDSCAPE |
`
`CeTY OF
`%
`
`=
`
`Incorporating novel approachesin the management of MDS
`.
`.
`beyond conventional hypomethylating agents
`5 O:
`= @ Olatoyosi Odenike
`
`we
`
`@o
`
`Department of Medicine, Section of Hematology/Oncology, The University of Chicago, Chicago, IL; and The
`University of Chicago Comprehensive Cancer Center, Chicago, IL
`
`In the last decade, the treatment of higher-risk myelodysplastic syndromes (MDS) has revolved around the azanu-
`cleosides, azacitidine and decitabine, which at lower doses are postulated to work predominantly via their effects on
`inhibition of DNA methyltransferases and consequent DNA hypomethylation. For patients whorelapse after, or do not
`respond to, hypomethylating agent therapy, the outcomeis dismal, and new agents and approachesthat have the
`potential to alter the natural history of these diseases are desperately needed. Allogeneic stem cell transplantis the only
`knownpotentially curative approach in MDS,butits applicability has been limited by the advanced age of patients and
`attendant comorbidities. There is now anincreasing array of new agents underclinical investigation in MDS that aim to
`exploit our expanding understanding of molecular pathways that are importantin the pathogenesis of MDS.This review
`focuses on a critical appraisal of novel agents being evaluated in higher-risk MDS that go beyond the conventional
`hypomethylating agent therapies approved by the US Food and Drug Administration.
`
` Limitations of treatment with HMAsare therefore obvious; namely,
`
`Learning Objectives
`
`e To understand the current treatment landscape in higher-risk
`MDSandthe limitations of conventional hypomethylating
`agent therapy
`e To gain an insightinto the novel agents and approaches under
`clinical investigation in higher-risk MDS
`
`manypatients have primary resistant disease, time to onset of response
`and achievement of best response can take several months, and
`myelosuppression before onset of response is nearly universal. Fur-
`thermore, despite the fact that a survival benefit has been demonstrated
`with azacitidine in higher-risk MDS,* these agents are not curative.
`The median duration of responseis in the 10- to 14-month range.***
`Outcomeafter failure of HMAsis particularly dismal, with a median
`survival of less than 6 months.’* Therefore, the development of new
`agents and strategies beyond the traditional HMAs approved by
`the FDA represents a significant area of unmet need at this time.
`This review will focus on novel agents and combinations under in-
`vestigation, including novel formulations of HMAs, novel epigenetic
`modulators, immunotherapeutic approaches, and therapies targeting
`specific molecular pathwaysin higher-risk myelodysplastic syndromes
`(Figure 1).
`
`Next-generation hypomethylating agents
`Because HMAsare S-phase-specific, a more prolonged exposure to
`the drug may allow greater incorporation into DNA.If used at rela-
`tively low doses, this would be hypothesized to lead to more sustained
`hypomethylation. A longer schedule of parenteral administration of
`decitabine and of azacitidine have been associated with significant
`activity in both acute myeloid leukemia (AML) and MDS,including
`poor prognosis subsets, lending somecredenceto that hypothesis.©?"!!
`These considerations, along with the very short half-lives (less than
`30 minutes) of conventional HMAs, coupled with the need for
`chronic administration to achieve or maintain a response, has spurred
`the developmentof the next generation of hypomethylating agents'”
`(Table 1). These include oral formulations of existing HMAsand/or
`
`Introduction
`The most notable development in the treatment of higher-risk myelo-
`dysplastic syndromes (MDS)in the last several years was the approval by
`the US Food and Drug Administration (FDA) of the hypomethylating
`agents (HMAs) 5-azacytidine (azacitidine) and 5-aza-2'deoxycytidine
`(decitabine) in 2004 and 2006,respectively. The use of these agents at
`lower doses, where their effects on DNA methyltransferase (DNMT)
`inhibition are postulated to predominate, results in objective responses
`including complete (CR) and partial (PR) responses in approximately
`15% to 20% ofpatients. An additional 20% to 30% achieve hematologic
`improvement (HI) in blood counts.’ Similar response rates have been
`demonstrated in clinical
`trials focused solely on higher-risk MDS
`(intermediate-2 or high-risk by International Prognostic scoring system
`[IPSS]). For example, in the landmark study by Fenaux etal, in which
`azacitidine was compared with conventional care regimens, the CR plus
`PRrate was 29%. The overall response rate (ORR) defined as CR, PR,
`and HI was 49%.* Responsesare gradual in onset, with a median onsetto
`response of 2 to 4 months and median time to best response of 5 to
`6 months. Responses can occur aslate as 12 months,although the majority
`of responses (>90%) would be expected to occur by 6 months™®)
`
`Conflict-of-interest disclosure: The authoris on the boardofdirectors or an advisory committee for Baxalta, CTI BioPharma, Celgene,Incyte, Pfizer, Jazz Pharmaceuticals,
`and ABIM, and hasreceived honoraria from AbbVie and Dava Oncology.
`Off-label drug use:| will be discussinginvestigational drugs in the treatment of MDSincluding novel formulations of azanuclesoides, novel epigenomic modulators,
`combinationsinvolving immune checkpointinhibitors, agents targeting specific genotypic subsets,kinase inhibitors, and BCL2inhibitors.
`
`460
`
`CELGENE 2102
`APOTEXv. CELGENE
`IPR2023-00512
`
`American Society of Hematology
`
`

`

`
`
`[= ——— —
`
`Transcription
`
`Immune
`Checkpoint
`Inhibition
`
`
`
`Figure 1. Novel agents and pathways underinvestigation in MDS. Epigenetic modulators including HMA, HDACis, BETinhibitors, and LSD1i affect
`chromatin structure and transcription; immune checkpointinhibition with a variety of monoclonal antibodies targeting the PD-1/PDL1 interaction, or
`CTLA-4 andits correspondingligand facilitate antigen (MHC) recognition by T-cell receptors; IDH1/2 inhibition affects the mutant enzyme within
`mitochondria, splicing modulation acts preferentially on cells harboring mutationsin splicing factors (splice mut); kinase inhibitors downregulate key
`signaling pathways including the RAS/MAPKand the PI3-K/AKT/m-TOR pathway.
`
`novel compounds,rationally designed, with a view to increasing or
`prolonging cellular exposure to HMAtherapy and ultimately im-
`proving therapeutic outcome.
`
`Oral azanucleosides
`In the last few years we have witnessed the introduction of oral
`azanucleosidesinto clinical trials. These agents may improvepatient
`convenience,eliminate injection site reactions, and facilitate chronic
`administration, including alternative dosing schedules, designed to
`lead to a more sustained cellular exposure.
`
`Oral azacitidine (CC-486) wasinitially studied in an open-label pilot
`trial. The agent was demonstrated to have 17% bioavailability, when
`compared with historical experience with parenteral azacitidine, after
`single-dose administration at 60 or 80 mg.'* A subsequentdose-finding
`study conducted in patients with MDS, chronic myelomonocytic
`leukemia (CMML), and AML, evaluating a 7 consecutive day oral
`administration schedule, established the maximum tolerated dose
`(MTD) of CC-486 as 480 mg daily for 7 days, with cycles being
`repeated every 28 days. Diarrhea was the dose-limiting toxicity. The
`most commonadverse events (AEs)included gastrointestinal toxicities,
`febrile neutropenia, and fatigue. ORR in patients with MDS or CMML,
`and without prior HMA exposure, was 73% (11 of 15 responded,
`including 6 CR and 5 HI). ORR in those whohadreceivedprior therapy
`was 35%.'* Extended dosing schedules of CC-486, 300 mgdaily for
`14 or 21 days, were investigated in lower-risk MDS.'* Accordingto
`the results of this early-phase trial, CC-486 is now being inves-
`tigated in a phase 3 trial (NCT01566695) in IPSS lower-risk MDS,
`with red cell transfusion dependency and thrombocytopenia. A recent
`analysis that focused on the experience of CC-486 across trials in
`
`patients who were previously exposed to HMA therapy showed that
`of 20 patients who had received 6 or more cycles of prior HMA
`therapy, 7 (35%) responded.'© Thus,there is an ongoingeffort evaluating
`CC-486 in the HMAfailure space, in combination with other novel
`approaches (Table 2).
`
`A majorhurdle in the clinical developmentoforal azanucleosidesis the
`fact that both azacitidine and decitabine are rapidly cleared by cytidine
`deaminase present in the gut and the liver, thus limiting their bio-
`availability. ASTX727, a novel formulation of oral decitabine paired
`with an oral cytidine deaminase inhibitor-E7727, is being studied in
`MDS,with a view to improvingthe bioavailability of the oral decitabine.
`Theresults ofa first-in-human phase 1 dose escalation trial of ASTX727
`demonstrated that the combination of the cytidine deaminase inhibitor
`E7727and oral decitabine, administered concurrently, successfully
`emulated the pharmacokinetic profile of intravenous (IV) decitabine.
`ASTX727 exhibited similar area under the curve parameters and a
`similar safety profile to IV decitabine, given at the standard dose and
`schedule.'’ The most common AEswere hematologic, including grade
`3 or greater thrombocytopenia, neutropenia, and febrile neutropenia.
`Nosignificant gastrointestinal-related AEs were reported. Preliminary
`report ofefficacy revealed a numberofresponses, including 5 CR and
`5 HI (n = 43). Four additional patients experienced a marrow CR.
`These results occurred in a patient population in which almosthalf had
`received prior HMA.
`
`A phase 2 fixed-dose confirmation stage of the study has just been
`completed, in whichpatients with intermediate- or high-risk MDS were
`randomly assigned in a crossoverdesignto receive the dose ofASTX727
`(35 mg decitabine plus 100 mg E7227)selected from the dose-escalation
`
`Hematology 2017
`
`461
`
`

`

`Table 1. Next-generation HMAsandinhibitors of other posttranscriptional/posttranslational marks
`
`Mechanism of action
`
`Phase of development
`
`Comments
`
`Intermediate 2/high-risk MDS-HMAfailure in
`phase 1, HMA-naive in phase 2
`Intermediate 2/high-risk MDS-HMAfailure
`
`MDS-HMAnaive; high orvery high-risk; stage 1,
`openlabel; stage 2, randomized, placebo
`controlled
`
`MDSexcludes low orvery low-risk disease,
`MDS/MPN, AML, myelofibrosis
`MDS-HMAfailure, AML-R/R
`MDS-HMAfailure; single-agent cohort or
`combination with azacitidine cohort
`
`+ATRA in MDS-R/R and AML-R/R
`
`+ATRA and L-DAC in MDS-HMAfailure and
`AML-R/R
`MDS or CMML HMAnaive with high or very
`high-risk and/or excess blasts
`Randomized, openlabel
`
`Reference
`
`NCT02103478
`
`NCT02907359
`
`_NCT03151304
`
`NCT02158858
`
`NCT02308761
`NCT02929498
`
`NCT02273102
`NCT02717884
`
`NCT02610777
`
`Agents
`
`ASTX727
`
`DNMTinhibition
`
`Phase 1/2
`
`Guadecitabine
`(SGI-1 10)
`Pracinostat azacitidine
`
`DNMTinhibition
`
`HDACinhibition DNMT
`inhibition
`
`CPI-0610
`
`BETinhibition
`
`RO6870810/TEN-010
`GSK2879552*
`
`BETinhibition
`LSD1 inhibition
`
`Tranylcypromine
`
`LSD1 inhibition
`
`Pevonedistat
`azacitidinet
`
`NAEinhibition
`
`DNMTinhibition
`
`L-DAC,low-dosecytarabine; R/R,relapsed and/orrefractory.
`*Notyet recruiting at time of manuscript submission.
`tAccrual is complete.
`
`Phase 3
`
`Phase 2
`
`Phase 1
`
`Phase 1
`Phase 2
`
`Phase 1
`Phase 1/2
`
`Phase 2
`
`phase of the study, versus IV decitabine at the standard dose and
`schedule. The phase 2 results confirm that the area underthe curve
`of oral ASTX727at this dose and schedule, as well as its effect on
`demethylation of repetitive (LINE-1) sequences, is similar to that
`observed with IV decitabine. Future plans for the development of
`this agent will likely involve further evaluation as an alternative to
`IV decitabine. Ideally, the clinical development of this agent and
`other oral azanucleosides should also include evaluation ofalternative
`
`doses and schedules targeted to induce more sustained hypomethylation
`and lower myelosuppression when compared with parenteral
`azanucleoside therapy.
`
`Preliminary evidence of promisingactivity in a phase 2 trial conducted
`exclusively in higher-risk previously untreated MDS or CMML was
`also recently reported.” Almosthalf the subjects enrolled (45%) had
`a complex karyotype, with 38% having TP53 mutations. Preliminary
`results indicate promising activity in this setting, with an ORR of
`61% (n = 36), including 28% with CR. Myelosuppression, requiring
`dose reduction, occurred in a third of patients. The most common
`nonhematologic AEs were grade1/2 nausea, fatigue, and dyspnea.
`These results suggest guadecitabine is worthy of further investigation
`in larger randomized trials in the frontline setting in MDS.
`
`Histone deacetylase inhibition
`Histone acetylation is a dynamic process, catalyzed by histone
`acetyltransferases, and is associated with an open chromatinstructure
`and recruitment to chromatin of factors involved in transcriptional
`regulation, DNArepair, and DNA replication. Histone deacetylases
`(HDACs) removeacetyl groups from thelysinetails of histones and
`lead to transcriptional repression and a closed chromatin configu-
`ration. HDACinhibitors (HDACis) were investigated on the premise
`that transcriptional de-repression associated with their use would
`result in upregulation of a variety of genes aberrantly silenced in
`cancercells, including tumor suppressor genes.” These agents have,
`however, been shownto affect both histone and nonhistoneproteins,
`and have beenassociated with pleiotropic effects on various genes
`involvedin cell cycle regulation, apoptosis, and angiogenesis. They
`have limited single-agent activity in myeloid malignancies.
`
`Rationally designed HMA formulations
`Anotherstrategy that has been employed to try to circumvent the rapid
`degradation of azanucleosides by cytidine deaminase is to develop
`anovelformulation that is chemically modified to berelatively resistant to
`deamination. Guadecitabine (SGI-110) is a novel dinucleotide of deci-
`tabine and deoxyguanosine,linked by a phosphodiester bond. Gradual
`cleavage of the phosphodiester bond is purported to lead to a slower
`release of the active decitabine moiety, thus prolonging cellular exposure
`to the drug. In a phase 1 study in patients with MDSand relapsed/
`refractory AML, myelosuppression was the dose-limiting toxicity.
`The MTD in MDSwasestablished to be 90 mg/m? administered
`daily (90 mg/m?/d) for 5 consecutive days. The biologically effective
`dose wassignificantly lower, and was determined to be 60 mg/m*/
`d for 5 consecutive days, based on the achievement of maximum DNA
`hypomethylationat this dose level.'* This dose/schedule (60 mg/m*/
`HDACishavebeeninvestigated extensively in combinationtrials with
`d) is now underfurther investigation in a variety of trials in AML,
`DNMTinhibitors (DNMTis), based on the hypothesis that therapeutic
`MDS, and CMML.In MDS or CMML,ongoing studies (Tables 1
`targeting of 2 pathwaysof epigenetic silencing in myeloid neoplasia
`and 2) have focused on the HMA failure space, evaluating single-agent
`would be synergistic. This hypothesized synergy between DNMTis
`guadecitabine in the phase 3 setting, or in combination with other
`and HDACis has been repeatedly demonstrated in vitro,”* but has
`novel agents in early-phase trials. These trials in the HMA failure
`been challenging to duplicate in vivo. Early-phase trials combining
`setting are based on preliminary results of encouraging activity seen
`HDACis and DNMTis confirmedthefeasibility of these combinations
`with guadecitabinein a phase2 trial, including patients with MDS who
`and yielded encouraging*”® results. This promise has, however, not
`have had prior exposure ttHMAs. '? Other groupsevaluating the agent
`
`in patients with disease that has relapsed after, or is refractory to, yetbeenrealized in the contextof a series of randomized phase2trials
`azacitidine therapy have reported more modestresults.”°
`in higher-risk MDSevaluating HDACi/DNMTi combinations®?””? vs
`
`462
`
`American Society of Hematology
`
`

`

`Table 2. Combination therapy with immune checkpoint inhibitors
`Phase of
`development
`
`Agents
`
`Mechanism ofaction
`
`Comments
`
`Reference
`
`Nivolumab
`ipilimumab
`azacitidine
`
`Immune checkpointinhibitors plus DNMT
`inhibition
`
`Phase 2
`
`Cohorts include single-agent immune
`checkpointinhibitors and combination
`with azacitidine; MDS-HMAfailure and
`naive
`
`NCT02530463
`
`Nivolumab
`azacitidine others*
`
`Immune checkpointinhibitors plus DNMT
`inhibition
`
`Phase 2/3
`
`Randomized phase 2, multiple experimental NCT03092674
`group study, selected exptal group in
`phase 2 proceeds to phase 3,
`azacitidine is the control group in both
`phase 2 and 3
`
`
`inhibition
`
`Durvalumab CC-486—Immune checkpoint inhibitors plus DNMT Phase 2 MDS-HMAfailure NCT02281084
`
`
`
`Durvalumab
`azacitidine
`
`Immune checkpointinhibitors plus DNMT
`inhibition
`
`Phase 2
`
`Pembrolizumab
`azacitidine*
`Nivolumab lirilumab
`azacitidine
`
`Immune checkpointinhibitors plus DNMT
`inhibition
`Anti-KIR MABplus immune checkpoint
`inhibitors + DNMTinhibition
`
`Phase 2
`
`Phase 2
`
`Randomized phase 2, MDS, HMAnaive-
`high or very highrisk or intermediate risk
`with excess blasts or poorrisk
`karyotype; AML = 65y old
`MDS-HMAfailure and naive cohorts,
`intermediate 1 or higherrisk
`Lirilumab + nivolumab in lower-risk MDS
`lirilumab + nivolumab + azacitidine in
`
`NCT02775903
`
`NCT03094637
`
`NCT02599649
`
`intermediate-2/high-risk MDS, HMAnaive
`Atezolizumab Single-agent immune checkpoint inhibitors|NCT02508870Immune checkpointinhibitors plus DNMT Phase 1
`
`
`azacitidine
`inhibition
`or combination with azacitidine. MDS-
`HMAfailure and HMA-naive cohorts
`
`
`
`Atezolizumab
`guadecitabine
`Ipilimumab
`decitabine
`
`Immune checkpointinhibitors plus DNMT
`inhibition
`Immune checkpointinhibitors plus DNMT
`inhibition
`
`Phase 1/2
`
`Phase 1
`
`NCT02935361
`
`NCT02890329
`
`MDS-HMAfailure, intermediate-1 or
`higher-risk
`MDS-HMAfailure and with excess blasts
`or MDS-relapsedafter allo-HCT, AML
`R/R, or =75 y old; allo-HCT naive and
`allo-HCTfailure cohorts
`
`
`inhibition
`
`Ipilimumab entinostat—Immune checkpoint inhibitors plus HDAC Phase 1b MDS-HMAfailure NCT02936752
`
`
`
`KIR, killer cell immunoglobulin-like receptor; MAB, monoclonal antibody.
`*Notyet recruiting at time of manuscript submission.
`
`single-agent DNMTi. A higher incidence of adverse events and/or
`early treatment discontinuation in the combination groups has been
`cited as an explanation for some of the recent disappointing results
`obtained in the context of these randomized trials.7*?? Pharmaco-
`dynamic antagonism has been cited as another potential explanation
`for failure of HDACi/DNMTi combinations to show benefit in the
`
`far. Ultimately, further developmentofthis class of drugs in MDSis
`predicated on being able to develop more tolerable combinationsthat
`are amenable to chronic administration and onthe ability to demonstrate
`a relative clinical advantage of the HDACi to these combination
`regimens.
`
`Lenalidomide-based combinations
`randomized setting. For example, the addition of entinostat did not
`Lenalidomide is FDA-approved for lower-risk MDSwith deletion 5q
`translate into clinical benefit, and less demethylation was observed in
`[del(5q)] MDS whoare transfusion dependent. The agent has more
`the combination group in the E1905 Intergroup randomized phase 2
`modestactivity in non-del (5q) lower-risk MDS (reviewed extensively
`trial, evaluating the HDACientinostat combined with azacitidine vs
`in an accompanying article by Giagounidis*’). In higher-risk MDS,
`single-agent azacitidine.® The possibility that this issue is schedule-
`lenalidomide was combined with azacitidine in a phase 1/2 trial in 36
`dependenthas been raised, with an overlapping schedule of admin-
`patients, with an ORR of 72%,including CR in 44% and HIin 28%.*!
`istration of the HDACi and DNMTileading to less incorporation of
`These promising results led to investigation of this combination in
`the azanucleoside into DNA,and consequently less hypomethylation.
`
`There is an ongoing randomized early-phasetrial in AML (NCT01305499) a larger groupofpatients with higher-risk MDS or CMMLinarecent
`designed to test this hypothesis by evaluating an overlapping vs
`North American Intergroup trial, $1117. This was a randomized
`a sequential schedule of administration of the azacitidine/entinostat
`phase 2 trial in which 277 patients were randomly assigned in a
`1:1:1 fashion to azacitidine combined with lenalidomide or with the
`combination.Atthis time, ongoing trials in MDSsuchas the azacitidine/
`pracinostat combination trial (Table 1) are focused on exploring al-
`ternative doses and/or schedules of HDACi/DNMTi combinations,
`with a view to improvingtolerability and outcome. In addition, beyond
`HDACi/DNMTitrials, other combinations involving HDACi and
`other novel agents such as immune checkpoint inhibitors are now
`underway in MDSin the HMA failure space (Table 2). At the moment,
`the future of HDACi-containing regimens in MDSis uncertain, given
`the multiple negative randomized trials that have been conducted thus
`
`HDACi vorinostat vs azacitidine monotherapy. The primary end-
`point was response rate. ORR wassimilar, at 38% for azacitidine
`monotherapy, 49% for azacitidine plus lenalidomide, and 27% for
`azacitidine plus vorinostat. Response duration and overall survival
`(OS) were also similar across treatment groups. There was a higher
`incidence of dose modifications and reductions in the combination
`
`groups compared with azacitidine monotherapy, implying poorer
`tolerability of the combination regimens.7®
`
`Hematology 2017
`
`463
`
`

`

`In patients with CMML,the ORR washigherin the azacitidine plus
`lenalidomide group, with 68% (13 of 19 patients) responding vs
`28% (5 of 18 patients) in the azacitidine monotherapy group.”
`Theseresults suggest the azacitidine plus lenalidomide combination
`may bebeneficial in patients with CMML. The numberofpatients
`with CMMLenrolled in the $1117 is too small, however, to be able
`to draw definitive conclusions. These results require validation in
`larger trials, focused on the CMMLpatient population.
`
`Novelinhibitors of other posttranslational or
`positranscriptional modifications
`Epigenomic dysregulation is a critical aspect of MDS pathogene-
`sis.** Beyondtargeting DNA methylation and HDACrecruitmentin
`MDS, however, there has been an increasing focus, in recent times,
`onthe clinical investigation of other inhibitors of posttranslational or
`posttranscriptional modifications that have the potential to affect the
`expression of key genes and pathways that are important in ma-
`lignant myeloid transformation.
`
`NEDD8-activating enzymeinhibition
`Pevonedistat is a NEDD8-activating enzyme (NAE) small molecule
`inhibitor. NAE regulates neddylation, which is a process by which
`Cullin-RING E3 ubiquitin ligases (CRLs) are activated and involves
`conjugation of the ubiquitin-like protein NEDD8to the Cullin protein
`scaffold. Activation of CRLsis, in turn, critical for proteasome-
`mediated protein degradation and proteasomal destruction of CRL
`substrates. Pevonedistat forms a covalent adduct with NAE, which
`leads to impaired CRL activation and accumulation of downstream
`CRL-dependentsubstrates. Several of these substrates are relevant to
`pathogenesis of myeloid malignancies, including cell cycle regulation,
`DNAdamage,andsignal transduction pathways. Preclinical work in
`AML demonstrated activity in cell lines, primary patient material, and
`murine xenograft models of AML.**
`
`Ina phase1 study of pevonedistatin relapsed refractory AML or MDS,
`modest single-agent clinical activity was observed: 17% ORR for
`schedule A (days 1, 3, and 5; n = 27) and 10% for schedule B (days1,
`4,8, and 11; n = 19). A subsequentdoseescalation trial was conducted
`investigating the combination of pevonedistat with azacitidine in
`treatment-naive AML. The MTD ofthe combination was pevonedistat
`20 mg/m? administered on days 1, 3, and 5 plus azacitidine 75 mg/m?
`administered on days 1 to 5, 8, and 9 on 28-day cycles. Grade 3
`hyperbilirubinemia and grade 4 aspartate aminotransferase (AST) were
`doselimiting. In the dose expansion phase ofthe study, of 55 patients
`enrolled, ORR was 60%, including 18 CR. Myelosuppression was
`common,with a febrile neutropenia rate of 25%. The median OS was
`7 months, with survival tending to be longer in patients with lowerblast
`burden below 30%.** A randomized trial is required to assess the
`relative contribution of pevonedistat to the combination. This is on-
`going in high-risk MDSandlow blast count AML(Table 1). Accrual to
`this trial was recently complete, and the results are eagerly awaited and
`are likely to determine the future developmentof this agent in MDS.
`
`Bromodomaininhibition
`Bromodomain and extraterminal (BET)proteins are epigenetic readers
`that recognize acetylatedlysinetails of histones, and thus areas of open
`chromatin structure or transcriptionally active sites. BET proteins
`possess conserved bromodomain modules that bind acetylated lysine
`tails and also interact with a numberofother proteins and function
`as scaffolds for molecules involved in gene transcription. BET
`proteins have been implicated in various cancers including myeloid
`
`malignancies. Inhibition of BET proteins leads to a significant re-
`duction of a numberof genes in a cell- and context-specific-dependent
`manner.*> Thefirst selective BET inhibitor, JQ1, was demonstrated to
`be active in vitro and in vivo in preclinical models of NUT midline
`carcinoma, a rare aggressive intrathoracic squamouscell carcinoma
`characterized by a rearrangement of the BET proteins BRD4 or
`BRD3, thusestablishing proof of concept for the therapeutic targeting
`ofBETproteins.*° In preclinical studies in AML,the use ofJQ] in AML
`cell lines and primary patient samples was associated with down-
`regulation of MYC and MYC-driven gene signatures specific to the
`leukemia stem cell population.*”°° There are a numberofclinical
`trials ongoing with BET inhibitors in various malignancies including
`MDS(Table 1). These trials are based largely on the potential
`promise of this class of drugs based on the experiencein preclinical
`models. It is too early at this juncture, however, to make definitive
`statements aboutclinical activity (andtolerability) of BETi in myeloid
`malignancies, including MDS.
`
`LSD1 inhibition
`Overexpression of the mono and dimethyl lysine demethylase, LSD1
`(also known as KDM1A)has been implicated in a variety of tumors
`including myeloid malignancies. LSD1 is important in maintaining
`embryonic stem cell pluripotency and regulates hematopoietic differ-
`entiation by keeping key differentiation genes and programssilenced.
`Inhibition of LSD1 or knockdownof the gene enhancesdifferentiation.
`LSD1 inhibition sensitized non-APL AML to all trans-retinoic acid
`
`(ATRA), and this was associated with an increase in histone 3 lysine 4
`dimethylation (H3K4me2), a marker of active transcription, and
`expression of myeloid differentiation genes. Furthermore, treatment
`with ATRA and a pharmacologic inhibitor of LSD1, tranylcypromine,
`resulted in a significant decrease in engraftment of primary AMLcells
`in nonobese diabetic-severe combined immunodeficient mice, sug-
`gesting this combination may target leukemia-initiating cells.*? Other
`novel LSD1 inhibitors have demonstrated activity in preclinical studies
`in AML and MDS“?Clinical trials are in progress evaluating LSD1
`inhibitors in combination with prodifferentiating agents such as ATRA
`or HMAsinpreviously treated patients with AML and MDS(Table 1).
`Theresults of these early-phase trials will determine thelikelihood for
`future developmentof these combinationsin larger groups of patients
`with higher-risk MDS.
`
`Immune checkpointinhibition
`Allogeneic stem cell transplant validates immunotherapy as a viable
`therapeutic strategy in MDS,but its applicability has been limited by
`the older age of patients at presentation and attendant comorbidities.
`The success of immune checkpoint inhibitors in solid tumors and
`Hodgkin lymphomahasled to the rapid introduction of these agents
`into clinical trials in other settings. These agents are based on the
`premise that a wide variety of tumors upregulate molecules such as
`PD-1/PDL-1 and CTLA4,which serve under normal circumstancesas
`“checkpoints” to recognize self and prevent autoimmunity.*! Cancer
`cells hijack these checkpoints as a meansto evade the immunesystem.
`Preclinical studies in myeloid malignancies have demonstrated that
`blockade of the PD-1/PDL1 pathway overcomes immuneevasion and
`prolongs survival in a murine model of AML.** Single-agentipili-
`mumab therapy has been investigated in a phase | trial in patients who
`relapsed after an allogeneic stem cell transplant and was associated
`with a CRin all 4 patients with extramedullary AML andin 1 patient
`with MDSthat had evolved to AML.*? These observations serve as
`proof of concept for immune checkpoint inhibition in AML/MDSin
`the postallogeneic stem cell transplant space.
`
`464
`
`American Society of Hematology
`
`

`

`Table 3. Other novel targeted agents and kinase inhibitors
`Phase of
`development
`
`Comments
`
`Reference
`
`Agents
`
`Mechanism of action
`
`Enasidenib (AG-
`221)*
`Ivosidenib (AG-
`120)*
`
`H3B8800 MDS-HMAfailure/intolerant, intermediate-2 or high-|NCT02841540Splicing modulator Phase 1
`risk; AML-R/R/U; CMML previously treated
`MDS- HMAfailure, intermediate-2 or high-risk, single- NCT02966782
`agent venetoclax and azacitidine+venetoclax
`combination cohorts
`
`IDH2 inhibition
`
`IDH1 inhibition
`
`
`
`Venetoclax
`azacitidine
`
`BCL2inhibition DNMT
`inhibition
`
`IDH2 mutant advanced and/or high-risk AML, MDS- NCT01915498
`RAEB1/2, or high-risk or R/R
`IDH1 R132 mutant advanced heme malignancy
`
`NCT02074839
`
`Phase 1/2
`
`Phase 1/2
`
`
`
`Phase 1
`
`Venetoclax
`azacitidine
`
`Rigosertib
`
`Ibrutinib
`azacitidine
`
`BCL2inhibition DNMT
`inhibition
`
`Phase 2
`
`Multitargeted kinase inhibition
`
`Phase 3
`
`BTKinhibition DNMTinhibition
`
`Phase 1b
`
`Selumetinib
`azacitidinet
`
`MEKinhibition DNMT
`inhibition
`
`Phase 1
`
`MDS- HMAnaive, intermediate-2/high-risk and > 5% NCT02942290
`blasts, randomized
`HMAfailure MDS-EB,includes RAEB-t; phase 3vs
`physician’s choice (includes best supportive care;
`azacitidine or DEC use also permitted)
`Intermediate or higher-risk MDS, HMAfailure (dose
`escalation stage only), HMAnaiveincluded in both
`stages of study
`Advanced myeloid malignancies including MDS-
`relapsed/refractory
`
`NCT02562443
`
`=NCT02553941
`
`BTK, Bruton tyrosine kinase; DEC, decitabine; MEK, mitogen-activated protein/extracellular signal-regulated kinase; RAEB,refractory anemia with excessblasts.
`*No longerrecruiting.
`tNotyet recruiting, clinicaltrials.org listing pending at the time of manuscript submission,
`
`Upregulation of PD-1 and PDL-1 expression has been demon-
`strated in primary MDS and AMLcells obtained from patients
`undergoing hypomethylating agent therapy, and has been linked to
`resistance to these agents.** Immune checkpoint inhibitor plus
`HMAcombinations in MDSare basedin part on the premise that
`HMAsmayact as an immunesensitizer*> and augmentthe activity
`of immune checkpoint blockade in MDSbyfacilitating recognition
`of malignant cells by cytotoxic CD8* T cells. Immune checkpoint
`blockade may also help overcome a potential mechanism ofre-
`sistance to azanucleoside therapy. The preliminary experience thus
`far suggests limited activity when immune checkpointinhibitors
`are used as single agents after HMAfailure.*° There are several
`combinationtrials of immune checkpoint inhibitors plus HMAsor
`HDACisthat are now ongoing in MDS, in both the HMA-naive and
`failure settings (Table 2). These trials are heterogeneous in design
`and patient population enrolled, and are largely predicated on the
`success of immune checkpoint inhibitors in solid tumors and
`Hodgkin lymphoma. Randomizedtrials will be required to decipher
`the relative contribution of immune checkpoint inhibition to these
`combinations.
`
`Therapies targeting specific genotypic subsets
`IDH1/2 inhibition
`Mutationsin isocitrate dehydrogenase enzymes (IDH1 and IDH2),are
`presentin approximately 15% to 20% ofpatients with AML. In MDS,
`these mutations are less common,being present in approximately 6%
`of cases, with the incidence rising with leukemic transformation.*7**
`Under physiologic conditions, IDH enzymescatalyze the conversion
`of isocitrate to a-ketoglutarate. IDH1/2 mutations confer a neomorphic
`enzymatic activity, resulting in isocitrate being converted to the
`oncometabolite R-2-hydroxyglutarate (2-HG). Elevated levels of
`2-HG result in competitive inhibition of a-ketoglutarate-dependent
`enzymesincluding TET2 and Jumonji-C enzymatic activity, leading
`to DNA andhistone hypermethylation, changes in chromatin con-
`figuration, and differentiation block.*? Small molecule inhibitors of
`mutant IDH1 or IDH2bindto the catalytically active site, preventing
`
`conversion of a-ketoglutarate to 2-HG andresulting in progressive
`reversal of histon

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