throbber
Liu J Hematol Oncol (2021) 14:49
`https://doi.org/10.1186/s13045-021-01062-w
`
`REVIEW
`Emerging agents and regimens for AML
`
`Open Access
`
`Hongtao Liu*
`
`
`
`Abstract
`Until recently, acute myeloid leukemia (AML) patients used to have limited treatment options, depending solely
`on cytarabine + anthracycline (7 + 3) intensive chemotherapy and hypomethylating agents. Allogeneic stem cell
`transplantation (Allo-SCT) played an important role to improve the survival of eligible AML patients in the past several
`decades. The exploration of the genomic and molecular landscape of AML, identification of mutations associated with
`the pathogenesis of AML, and the understanding of the mechanisms of resistance to treatment from excellent transla-
`tional research helped to expand the treatment options of AML quickly in the past few years, resulting in noteworthy
`breakthroughs and FDA approvals of new therapeutic treatments in AML patients. Targeted therapies and combina-
`tions of different classes of therapeutic agents to overcome treatment resistance further expanded the treatment
`options and improved survival. Immunotherapy, including antibody-based treatment, inhibition of immune negative
`regulators, and possible CAR T cells might further expand the therapeutic armamentarium for AML. This review is
`intended to summarize the recent developments in the treatment of AML.
`Keywords: AML, Targeted therapy, Novel treatment
`
`Introduction
`AML is a heterogeneous disease, defined by a broad
`spectrum of genomic changes and molecular mutations
`that influence clinical outcomes and provide potential
`targets for drug development. The updated 2017 Euro-
`pean LeukemiaNet (ELN) risk stratification guidelines
`combining cytogenetic abnormalities and genetic muta-
`tions have been widely used to predict the prognosis of
`AML patients [1], while others have been exploring to
`incorporate additional prognostic factors into ELN-2017
`guidelines to improve the risk stratification models [2].
`Advanced by basic and translational research, espe-
`cially through large scale genomic analysis to understand
`the molecular landscape of AML, the development of tar-
`geted therapies, such as targeting fms-like tyrosine kinase
`3 (FLT3) and isocitrate dehydrogenase 1 and 2 (IDH1
`and IDH2) mutations, the treatment of AML landscape
`changed significantly with FDA approvals for several
`
`*Correspondence: hliu2@medicine.bsd.uchicago.edu
`Section of Hematology/Oncology, Department of Medicine, The
`University of Chicago Medical Center, 5841 S. Maryland Ave, MC 2115,
`Chicago, IL 60637-1470, USA
`
`new drugs in the past several years. Even with all these
`improvements, primary resistance to initial treatment
`and disease relapse remain huge unmet need in the treat-
`ment of AML. The majority of AML patients still even-
`tually succumb to the disease. We still have a long way
`to further improve the survival of the AML patients, thus
`many investigational drugs have been explored to target
`the primary and secondary treatment resistance in AML
`patients.
`This review will provide updates of the emerging thera-
`peutic approaches for the treatment of AML, including
`combinations with mutation driven targeted treatments,
`novel immunotherapies in the myeloid disease.
`
`Targeted therapies: alone or combination
`BCL-2 inhibitor: venetoclax
`BCL-2 is a member of the BCL-2 family of anti- and pro-
`apoptotic proteins. BCL-2 protects cells against apopto-
`sis. BCL-2 expression in AML has been associated with
`decreased sensitivity to cytotoxic chemotherapy and a
`higher rate of relapse [3]. Venetoclax is an orally bioavail-
`able selective inhibitor of BCL-2, promoting intrinsic
`apoptotic pathway activation resulting in mitochondrial
`
`© The Author(s) 2021. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which
`permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the
`original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or
`other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line
`to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory
`regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this
`licence, visit http:// creat iveco mmons. org/ licen ses/ by/4. 0/. The Creative Commons Public Domain Dedication waiver (http:// creat iveco
`mmons. org/ publi cdoma in/ zero/1. 0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
`
`CELGENE 2095
`APOTEX v. CELGENE
`IPR2023-00512
`
`

`

`Liu J Hematol Oncol (2021) 14:49
`
`Page 2 of 20
`
`outer membrane permeability through dissociation of
`BCL-2 mediated sequestration of BH3 proteins BIM and
`BID and effector proteins BAX and BAK. Venetoclax was
`initially approved by U. S. Food and Drug Administration
`(FDA) in 2016 to treat individuals with chronic lympho-
`cytic leukemia (CLL) with deletion (17p).
`Venetoclax + hypomethylating agents or low dose cytarabine
`Early studies using venetoclax as monotherapy in AML
`demonstrated only modest efficacy in high-risk relapsed/
`refractory (R/R) AML patients with an overall response
`rate (ORR) of 38% and complete remission/complete
`remission with incomplete hematologic recovery (CR/
`CRi) of 19%. The responses were short lived, with overall
`survival (OS) of only 4.7 months [4]. Based on promis-
`ing results from two large Phase 1b/II trials using com-
`bination of a hypomethylating agent (HMA) or low-dose
`cytarabine (LDAC) with venetoclax in untreated older
`AML patients [5, 6], FDA granted accelerated approval
`to venetoclax in combination with azacitidine (AZA) or
`decitabine (DEC) or LDAC for the treatment of newly-
`diagnosed (ND) AML in adults who are age 75 years or
`older, or who have comorbidities that preclude use of
`intensive induction chemotherapies in 2018.
`Recently published Phase III randomized studies con-
`firmed the results from these early single arm trials, and
`demonstrated a significant survival benefit from add-
`ing venetoclax to azacitidine and to LDAC [7, 8]. The
`major findings from the VIALE-A and VIALE-C trials
`are summarized in Table  1. In summary, the VIALE-A
`trial included 431 patients without history of exposure
`to azacitidine. At a median follow-up of 20.5  months,
`
`the median OS was 14.7 months in the azacitidine-vene-
`toclax group and 9.6  months in the control group. The
`incidence of CR and composite complete remission rate
`(cCR) (CR + CRi) were significantly higher with azaciti-
`dine-venetoclax than with the control regimen. How-
`ever, there were higher rates in key adverse events in the
`azacitidine-venetoclax group than those in the control
`group, but they were manageable [7]. The VIALE-C study
`assigned 211 patients to either venetoclax (n = 143) or
`placebo (n = 68) in 28-day cycles, plus LDAC on days 1
`to 10. In contrast to VIALE-A trial, 20% enrolled patients
`had received prior HMA treatments. The planned pri-
`mary analysis showed a 25% reduction in risk of death
`with venetoclax plus LDAC vs LDAC alone, although
`this was not statistically significant. Median OS was 7.2
`vs 4.1 months, respectively. Unplanned analyses with an
`additional 6-months follow-up demonstrated median
`OS of 8.4 months for the venetoclax arm (HR, 0.70; 95%
`CI, 0.50–0.98; P = 0.04). CR/CRi rates were 48% and 13%
`for venetoclax plus LDAC and LDAC alone, respectively.
`Thus, venetoclax plus LDAC demonstrated clinically
`meaningful improvement in remission rate and OS vs
`LDAC alone, with a manageable safety profile [8]. Based
`on these confirmatory data, FDA granted full approval to
`these venetoclax combinations for treating newly diag-
`nosed AML patients. Both trials established new stand-
`ard of care for unfit newly diagnosed AML patients. Since
`VIALE-A trial excluded patients with previous exposure
`to azacitidine, and 20% patients enrolled on the VIALE-C
`trial had exposure to HMA, venetoclax plus LDAC might
`be a preferred consideration for patients who received
`HMAs in the past.
`
`Table 1 Comparison of randomized prospective studies on venetoclax-based combinations
`LDAC + venetoclax
`Regimen
`
`AZA + venetoclax
`
`in AML: AZA + venetoclax vs
`
`LDAC + venetoclax
`
`Phase
`Population
`Control arm
`h/o HMA
`Patient number
`
`Median age
`(range), years
`30-day mortality, %
`cCR (CR) rate, %
`MRD negativity, %
`Time to CR (response)
`Median DOR, months
`Median OS, months
`Reference
`
`III VIALE-A trial
`Age > 75 years or unfit for chemotherapy
`AZA
`No
`431
`(286 in AZA + venetoclax)
`76 (49–91)
`
`III VIALE-C trial
`
`LDAC
`Yes, allowed (20%)
`211
`(143 in LDAC + venetoclax)
`76 (36–93)
`
`7%
`66.4% (36.7%)
`N/A
`1.3 months (0.6–9.9)
`17.5 (13.6 to NR)
`14.7 (11.9–18.7)
`[7]
`
`13%
`48% (27%)
`6%
`N/A most response at the end of cycle 2
`NA
`8.4 (5.9–10.1)
`[8]
`
`

`

`Liu J Hematol Oncol (2021) 14:49
`
`
`Page 3 of 20
`
`Both trials also identified that patients with NPM1
`and IDH1/2 mutations had high CR rates of 91%, and
`71%, respectively with HMA + venetoclax [5] and high
`CR/CRi rates (89% and 72%), respectively, when treated
`with venetoclax + LDAC [6]. Patients with FLT3 muta-
`tions (Internal tandem duplication (ITD) and/or tyrosine
`kinase domain (TKD) also demonstrated high CR rate
`of 72% [5]. On the other hand, inhibitors to these muta-
`tions have been developed and will be discussed in the
`following sections. It would be continued debate on how
`to choose the first line treatment for AML with these
`mutations: hypomethylating agents with IDH1/2 inhibi-
`tors vs venetoclax-based combination; how to sequence
`the treatment options: venetoclax-based combinations
`first followed by IDH1/2 inhibitors at disease relapse/
`progression or the other way around; or use three drugs
`combination with HMA + venetoclax + IDH1/2 inhibi-
`tor to get deeper remission. Only randomized clinical
`trials could eventually answer these important clinical
`questions.
`Venetoclax + intensive chemotherapy
`Not surprisingly, venetoclax has been studied in com-
`binations with intensive chemotherapy as well (summa-
`rized in Table  2). A retrospective report of 13 patients
`treated with FLAVIDA salvage therapy (fludarabine,
`cytarabine, and idarubicin in combination with veneto-
`clax 100 mg daily for 7 days; dose reduced due to con-
`current azole administration) compared to a control
`cohort received FLA-Ida (fludarabine, cytarabine, and
`idarubicin) reported a higher but not statistically sig-
`nificant CR/CRi rate of 69% compared to 47% in the
`control cohort [9]. A phase 1b/II trial of medically fit
`patients with R/R AML receiving FLAG-Ida induction
`
`and consolidation in combination with a 14 days course
`of venetoclax was conducted at MD Anderson. Early
`results were promising with CRc of 74% in all the patients
`and an impressive CRc of 91% in newly diagnosed (ND)
`patients. Consistent with known venetoclax resist-
`ance mechanisms, high levels of MCL-1 expression
`were found in patients who relapsed following FLAG-
`Ida + venetoclax [10]. The updated data of 62 patients
`(27 with ND AML and 35 with R/R AML) from the trial
`was recently presented. The ORR was 84%, with 89% of
`ND AML and 66% of R/R AML patients achieving a CRc.
`83% of patients achieved minimal residual disease (MRD)
`negative (MRD-) status assessed by flow cytometry. After
`a median follow up of 11  months, median OS was not
`reached. The addition of venetoclax to FLAG-ida demon-
`strated robust efficacy with acceptable safety profile [11].
`The CAVEAT study reported data on 51 newly diag-
`nosed patients with AML, either de novo or second-
`ary, who were treated in five venetoclax dose-escalation
`cohorts (50–600 mg; venetoclax was given over 14 days,
`day -6 to 7 with induction chemotherapy (cytarabine
`100  mg/m2 days 1–5 and idarubicin 12  mg/m2 intrave-
`nously days 2–3)). The same venetoclax dose and sched-
`ule was given for four cycles of consolidation (cytarabine,
`days 1–2, and idarubicin, day 1), and as maintenance
`(up to seven 28-day cycles). The overall CR/CRi rate was
`72%, but was 97% in the 28 patients with de novo AML
`and only 43% in secondary AML. [12]. In our center, we
`have used HiDAC + mitoxantrone + venetoclax for sev-
`eral heavily pretreated patients with R/R acute leukemia
`to control the disease prior to allogeneic stem cell trans-
`plantation (allo-SCT) (personal experience). This combi-
`nation warrants further study in both newly diagnosed
`and R/R AML setting.
`
`Table 2 Summary of venetoclax-based combinations in AML
`
`Combination
`
`Phase
`
`Disease status
`
`Patient number
`
`CR/CRi rate, %
`
`References
`
`FLA-Ida
`FLAG-ida
`CAVEAT (5 + 2)
`
`DEC10
`
`CLIA
`CLAD/LDAC, alternating
`with AZA
`CPX-351
`
`CPX-351 LIT
`GO
`
`Retrospective
`Ib/II
`
`Ib
`
`II
`
`II
`II
`
`II
`
`Ib
`Ib
`
`R/R AML
`ND AML
`R/R AML
`ND AML
`
`ND AML
`R/R AML
`ND AML
`ND AML
`
`R/R AML
`ND AML
`ND AML
`R/R AML
`
`13
`27
`35
`51
`
`70
`55
`18
`48
`
`69%
`89% in ND AML
`66% in R/R AML
`72% in all
`97% in de novo AML
`43% secondary AML
`86% in ND AML
`42% in R/R AML
`88%
`94%
`
`17
`1
`44 planned
`24 planned
`
`37%
`
`NA
`NA
`
`[9]
`[10, 11]
`
`[12]
`
`[13]
`
`[14]
`[15]
`
`[16]
`
`[17]
`[18]
`
`

`

`Liu J Hematol Oncol (2021) 14:49
`
`Page 4 of 20
`
`The results of ten-days of decitabine (DEC10) with
`venetoclax (DEC10-VEN) in AML and high-risk MDS
`were reported. DEC10-VEN is safe and highly effec-
`tive in newly diagnosed AML and can serve as an
`effective bridge to SCT. Median OS in treatment naïve
`AML patients who subsequently underwent SCT
`was not reached (1  year OS of 100%). For previously
`treated AML patients, OS was 22.1  months [13]. In
`addition, propensity score matched analysis (PSMA)
`was employed to compare outcomes of 54 younger
`adult patients with R/R AML treated on the prospec-
`tive phase 2 trial of 10-day decitabine and venetoclax
`(DEC10-VEN) with a historical cohort of patients
`treated with intensive chemotherapy. The analysis
`demonstrated that DEC10-VEN provided comparable
`response of CR/CRi, OS, and rate of patient to proceed
`SCT to non-venetoclax based intensive chemotherapy.
`Thus, DEC10-VEN represents an appropriate salvage
`therapy, and provides an appropriate backbone for add-
`ing novel therapies in R/R AML patients [19].
`The addition of venetoclax to cladribine, idarubicin,
`and Ara C (CLIA) was safe and effective in ND patients
`with AML. The combination was not associated with
`early mortality or prolonged myelosuppression, but
`did result in high rates of durable MRD negative remis-
`sions (NCT02115295) [14]. Addition of venetoclax to a
`low-intensity backbone of cladribine + LDAC (CLAD/
`LDAC) alternating with HMA for older patients with
`newly diagnosed AML provided a CR/CRi rate of 94%;
`and among the subset of patients who had CR with
`complete count recovery, the MRD negative rate was
`92%. The regimen was well tolerated, with 4-week mor-
`tality rates of 0%. With a median follow-up of more
`than 11 months, the median OS has not been reached
`(NR), with 12-month OS rates of 70% [15]. Full dose
`CPX-351 plus 7  days of VEN (300  mg on D2-8) was
`demonstrated to be tolerable with acceptable toxici-
`ties in patients with R/R AML with an ORR of 44%; and
`ORR was high at 60% in patient without prior VEN
`exposure, compared to just 17% among those who had
`prior VEN. 86% of responding patients proceeded to
`SCT. The median OS overall was 6.4 months; and the
`median OS was not reached among the responders
`[16].
`Other ongoing trials include open-label, multicenter,
`2-part, phase 1b study (NCT04038437) to determine
`the maximum tolerated dose and evaluate the safety,
`efficacy, and pharmacokinetics of CPX-351 lower-inten-
`sity therapy (LIT) plus venetoclax [17]. Another single
`arm, open-label, multi-center, dose-escalation phase Ib
`study is evaluating the combination of venetoclax and
`gemtuzumab ozogamicin in R/R CD33 + AML patients
`(NCT04070768) [18].
`
`Venetoclax + experimental drugs or targeted inhibitors
`Given the proven synergies of BCL-2 inhibition, mul-
`tiple combinations with targeted agents, and veneto-
`clax are under investigation. There are many ongoing
`combinations of therapies targeting BCL-2 and other
`pathways, including FLT3 inhibitors (gilteritinib) and
`IDH1 and 2 inhibitors (Ivosidenib and enasidenib)
`(will be discussed in the later sections), MCL-1 inhibi-
`tors (VU661013, A-1210477); MEK1/2 inhibitor (cobi-
`metinib), and MDM2 inhibitor (idasanutlin) (reviewed in
`[20]), combination with TKI in Ph + acute leukemia [21]
`and other emerging pre-clinical combinations includ-
`ing small-molecule inhibitors of CDK9 (the orally active
`A-1592668 and the related analog A-1467729) leading
`to down-expression of MCL-1 [22]; the Exportin inhibi-
`tor, Selinexor, [23]; BET inhibitors, ABBV-075, [24]; SRC
`family kinases (SFK) and Bruton’s tyrosine kinase (BTK)
`inhibitor, ArQule 531 (ARQ 531), [25]; and it is expecting
`much more novel combinations to come.
`
`Resistance mechanisms
`HMA + venetoclax or LDAC + venetoclax have clearly
`advanced the treatment of AML for older or unfit AML
`patients. Unfortunately, these regimens are unlikely
`to provide cure as most patients have relapsed at the
`median of 7 cycles of treatment. A retrospective study
`demonstrated that the outcome of 41 patients who failed
`to respond to HMA + venetoclax was very poor with the
`median OS of only 2.4  months despite salvage therapy
`[26]. To understand the resistance mechanisms, DiNardo
`CD et al. analyzed 81 patients receiving these venetoclax-
`based combinations to identify molecular correlates of
`durable remission, initial response followed by relapse
`(adaptive resistance), or refractory disease (primary
`resistance). Acquisition or enrichment of clones with
`activation of the signaling pathways such as FLT3 or RAS
`or bi-allelic mutations perturbing TP53 were most com-
`monly identified among primary and adaptive resistance
`to venetoclax-based combinations. Single-cell studies
`identified heterogeneous and sometimes divergent inter-
`val changes in leukemic clones within a single cycle of
`therapy, highlighting the dynamic and rapid occurrence
`of therapeutic selection in AML. In functional studies,
`gain of FLT3-ITD mutation or loss of TP53 conferred
`cross-resistance to both venetoclax and cytotoxic-based
`therapies [27]. These data confirmed the previous find-
`ings that TP53 apoptotic network is the primary media-
`tor of resistance to BCL-2 inhibition in AML cells [28].
`Interestingly, recent study demonstrated that monocytic
`AML is intrinsically resistant to venetoclax + AZA due to
`loss of expression of the venetoclax target of BCL-2, but
`instead preferentially reliant on MCL-1 for the survival.
`
`

`

`Liu J Hematol Oncol (2021) 14:49
`
`
`Page 5 of 20
`
`Thus, venetoclax + AZA treatment selects monocytic
`disease at disease relapse, which is derived from pre-
`existing monocytic subclones [29]. AML patients with
`monocytic disease or TP53 mutation might have high
`risk to be resistant to venetoclax-based combinations,
`and clinical trials targeting TP53 mutation or trials spe-
`cifically targeting monocytic AML might be considered
`over venetoclax-based combinations.
`Future clinical research will focus on deepening the
`responses provided by HMA + venetoclax with addi-
`tional targeted agents, like ivosidenib in IDH1 mutated
`AML (to be discussed in next section), FLT3 inhibitors,
`and novel pathways inhibitors to eventually cure a greater
`fraction of newly diagnosed AML, and to explore new
`strategies to deal with relapses after venetoclax-based
`therapies.
`
`IDH1/2 inhibitors
`IDH1 and IDH2 are critical enzymes for the oxidative car-
`boxylation of isocitrate. A mutation in one of these genes
`results in increased concentration of 2-hydroxyglutarate
`(2-HG). 2-HG causes DNA and histone hypermethyla-
`tion, leading to blocked cellular differentiation and tumo-
`rigenesis. Mutations in IDH1 or IDH2 are present in 5%
`to 15% and 10% to 15% of patients with newly diagnosed
`AML, respectively [30]. Oral, small-molecule inhibitors
`have been developed for both mutant IDH1 (ivosidenib)
`and IDH2 (enasidenib). In R/R AML, ivosidenib and
`enasidenib as single agent produced promising responses
`for the corresponding mutations with ORR of 41.6% (CR:
`21.6%) with median OS of 8.8 months [31] and ORR of
`40.3% (CR 20.6%) with median OS of 9.3  months [32]
`respectively. FDA approved ivosidenib and enasidenib
`for patients with relapsed or refractory IDH1 and IDH2
`mutated AML, respectively, in 2018. In the front line
`setting, both inhibitors have also demonstrated clini-
`cal effectiveness [33, 34], leading to FDA approval of
`ivosidenib for patients with newly diagnosed IDH1
`mutated AML based on an ORR of 42% (CR: 30%) with
`median OS of 12.6 months in older patients not eligible
`for intensive therapy [34].
`The Phase 3 IDHENTIFY study evaluating enasidenib
`plus best supportive care (BSC) versus conventional
`care regimens, which included BSC only, azacitidine
`plus BSC, low-dose cytarabine plus BSC, or intermedi-
`ate-dose cytarabine plus BSC, did not meet the primary
`endpoint of OS in patients with R/R AML with an IDH2
`mutation. The safety profile of enasidenib was consistent
`with previously reported findings. IDH inhibitors alone
`are unlikely to provide cure or durable remission for R/R
`AML, but they might provide excellent disease control
`with low toxicity and a bridge to allo-SCT.
`
`IDH inhibitors work in part through induction of
`differentiation of malignant cells, leading to differen-
`tiation syndrome in 10% to 20% of patients. Clinical
`features are similar to those seen in patients with acute
`promyelocytic leukemia (APL) treated with ATRA-
`based regimens [35, 36]. Early studies established a firm
`association between IDH mutations and serum 2-HG
`concentration in AML, and confirmed that serum
`oncometabolite measurements provide useful diag-
`nostic and prognostic information that can improve
`patient selection for IDH-targeted therapies [37]. How-
`ever, 2-HG level reduction and clearance of IDH muta-
`tion by next generation sequencing (NGS) assay does
`not correlate with the clinical response. These inhibi-
`tors are unlikely to provide cure of the AML due to pri-
`mary resistance from co-mutations in other pathways
`especially the NRAS/KRAS, and MAPK pathway effec-
`tors PTPN11, NF1, FLT3 and others [38] and secondary
`resistance from development of second-site IDH2 mis-
`sense mutations or isoform switching [39, 40].
`Since IDH1/2 mutations lead to DNA and histone
`hypermethylation, HMAs might have synergistic effects
`in combination of IDH inhibitors. Combination of HMAs
`with IDH inhibitors has been studied. The combination
`of ivosidenib and azacitidine was studied in 23 patients
`with IDH1 mutated AML as front line treatment. The
`ORR was 78% with CR/CRh rate of 70%, and median
`time to response of 1.8 months; median response dura-
`tion was not yet reached. The ivosidenib and azacitidine
`combination was well tolerated with a safety profile con-
`sistent with ivosidenib or AZA monotherapy and with
`17% incidence of IDH differentiation syndrome. Clear-
`ance of mutated IDH1 was seen in 63% patients with CR/
`CRh. CR and ORR rates exceeded those expected from
`AZA alone [41]; 83% CR/CRh patients achieved MRD
`negativity by flow cytometry [42]. AGILE, a global, dou-
`ble-blind, randomized, placebo-controlled, phase III trial
`for patients with previously untreated IDH1 mutated
`AML who are not candidates for intensive therapy
`(NCT03173248) is actively enrolling patients from 172
`study centers across the world [43]. Patients are ran-
`domly assigned to AZA + ivosidenib or AZA + placebo.
`As for the IDH2 inhibitor of enasidenib, the phase II
`portion of an open-label, randomized phase I/II study
`of enasidenib (E) + AZA (“E + A”) vs AZA monother-
`apy (“A”) in patients with mutated IDH2 (mIDH2) ND
`AML (NCT02677922) was recently reported [44]. 101
`patients with intermediate- or poor-risk cytogenet-
`ics were randomized 2:1 to E + A or A in 28-day cycles.
`ORR (71% vs 42%) and CR (53% vs 12%) rates were sig-
`nificantly improved with E + A with greater clearance of
`mIDH2 allele frequency. Time to first response was about
`2 months in each arm and the time to CR was 5.5 months
`
`

`

`Liu J Hematol Oncol (2021) 14:49
`
`Page 6 of 20
`
`(range, 0.7–19.5). There was no difference in median PFS
`and OS so far [44].
`As discussed in the section of Azacitidine and vene-
`toclax, this combination is very effective in patients
`with IDH1/2 mutation. In a pooled retrospective study,
`79 patients with IDH1/2 mutation were identified and
`treated with VEN + AZA on either the Phase Ib or the
`randomized Phase III (VIALE-A) trials. CR/CRh was
`72% (95% CI: 61%-82%) in the whole population. In
`patients with IDH1, CR/CRh was 59%, median time
`to first CR/CRh response was 2.3  months, and median
`duration of response (DOR) and OS were 21.9 (7.8–29.5)
`months and NR. In patients with IDH2, CR/CRh rates
`were 80%, median time to first CR/CRh response was
`1.0  month. Median DOR and median OS (mOS) were
`NR. Thus, VEN + AZA provided high response rates,
`long DOR, and mOS among treatment-naïve patients
`with IDH1/2 mutation ineligible for intensive chemo-
`therapy with acceptable safety profile [45]. As mentioned
`previously, it will be a continued debate to optimize the
`front line treatment for unfit AML patients with IDH1/2
`mutations.
`There is also a rationale for combining IDH inhibitors
`with BCL-2 inhibitors, since the accumulation of 2-HG
`caused by IDH mutations could decrease the mitochon-
`drial threshold for induction of apoptosis induced by
`BCL-2 inhibition with venetoclax [46]. The combina-
`tion therapy of ivosidenib (IVO) plus venetoclax (VEN)
`with or without azacitidine was found to be effective
`against AML harboring an IDH1 mutation in a phase
`Ib/II trial [47]. Patients with AML or high-risk MDS
`were assigned to one of three cohorts, either receiving
`IVO + VEN 400 mg, IVO + VEN 800 mg, or IVO + VEN
`400  mg + AZA. The median time to best response was
`2  months. In 18 evaluable patients, cCR rate was 78%
`overall (treatment naive: 100%; R/R: 75%), and 67%, 100%,
`and 67% by cohort with median time to best response of
`
`2 months. IVO + VEN + AZA therapy was well tolerated
`and highly effective for patients with IDH1 mutated AML
`[47]. It is reasonable to expect that combination of the
`IDH2 inhibitor enasidenib with venetoclax and azaciti-
`dine might also provide better outcomes than enasidenib
`alone or enasidenib with azacitidine, since enasidenib
`plus venetoclax demonstrated superior anti-leukemic
`activity against IDH2 mutated AML in patient-derived
`xenograft models [48]. Table 3 summarizes the trials of
`combinational therapies for newly diagnosed unfit AML
`patients with IDH1/2 mutation.
`
`Targeting FLT3 mutations
`FLT3 Mutations occurs in approximately 30% patients
`with newly diagnosed AML (20% to 25% with FLT3-ITD
`mutation, 5% to 10% with FLT3-TKD), and associates
`with more proliferative disease, increased risk of relapse,
`and inferior survival. Randomized phase III RATIFY
`study led to approval of 7 + 3 + midostaurin as front
`line for young fit patients [51], and randomized phase
`III ADMIRAL study with single-agent gilteritinib estab-
`lished the approval of gilteritinib for the treatment of R/R
`FLT3-mutated AML [52]. The phase III randomized trial
`using quizartinib vs investigator choice salvage chemo-
`therapy in patients with R/R FLT3-ITD mutated AML
`met the primary objective of OS improvement [53], lead-
`ing its approval in Japan, but not in the US. Many studies
`to combine these FLT3 inhibitors are under investigation.
`An open-label, phase 1 study (NCT02236013) assessed
`the safety/tolerability and anti-leukemic effects of gilteri-
`tinib plus 7 + 3 induction, consolidation, and main-
`tenance therapy in fit adults with newly diagnosed
`FLT3-mutated AML. 80 patients with median age of
`59  years were allocated to treatment. The maximum
`tolerated dose of gilteritinib was 120 mg daily. CRc was
`achieved by 81.8% of patients across all dose groups with
`mutational clearance (FLT3 ITD signal ratio of ≤ 10–4
`
`Table 3 Summary of combination of targeted-therapy trials in IDH1/2-mutant newly diagnosed AML
`
`Regimens
`
`Phase
`
`Patient
`Number
`
`CR/CRi rate, %
`
`Time to CR or response
`(median), months
`
`OS (median),
`months
`
`References
`
`HMA + venetoclax
`AZA + venetoclax
`AZA + venetoclax
`LDAC + venetoclax
`(HMA naïve)
`AZA + ivosidenib
`AZA + enasidenib
`Venetoclax + ivosidenib
`AZA + venetoclax + ivosidenib
`
`Ib
`III
`Pooled data from
`two trials
`Ib/II
`III
`Ib
`II
`Ib/II
`Ib/II
`
`35
`46
`79
`
`18
`
`23
`68
`12
`6
`
`71
`75.4
`72
`
`72
`
`69.6
`68
`83
`67
`
`2.1
`N/A
`1.0
`
`1.4
`
`3.7
`5
`NA
`NA
`
`24.4
`N/A
`24.5
`
`19.4
`
`N/A
`22.0
`NA
`NA
`
`[5]
`[7]
`[45]
`
`[6, 8]
`
`[49]
`[50]
`[47]
`[47]
`
`

`

`Liu J Hematol Oncol (2021) 14:49
`
`
`Page 7 of 20
`
`after induction or consolidation) was achieved by 70% of
`patients with FLT-ITD mutation receiving a gilteritinib
`dose of ≥ 120 mg [54]. Two large randomized clinical tri-
`als of induction and consolidation chemotherapy plus
`gilteritinib vs midostaurin in FLT3 mutated AML patients
`are ongoing in the US (PrECOG trial) (NCT03836209))
`and in Europe (HOVON 156 AML / AMLSG 28–18 trial
`(NCT04027309)).
`The LACEWING study is a phase 3 trial to randomize
`FLT3 mutated ND AML patients ineligible for intensive
`induction chemotherapy to get gilteritinib plus azaciti-
`dine vs azacitidine alone. The safety cohort enrolled 15
`patients and established dose of gilteritinib of 120 mg to
`be used in combination with azacitidine. Overall, a CRc
`of 67% was observed with median duration of remission
`of 10.4 months for the CRc responders. The combination
`treatment was well tolerated with no unexpected adverse
`effect [55]. While the data provides a promising option
`of gilteritinib plus azacitidine for newly diagnosed FLT3-
`mutated unfit AML patients, the company announced
`that Phase 3 LACEWING trial failed to meet primary
`end point of OS at a planned interim analysis and the
`study was terminated for futility in December 2020.
`Many lessons have been learned in the AML field that
`high response rate in AML will not necessary transform
`into survival benefit.
`In the R/R setting, a phase 1b study tested the safety
`and efficacy of combining venetoclax at 400  mg with
`gilteritinib at 120  mg daily. 39 patients were enrolled,
`and among them 64% had previous history of FLT3
`TKI exposure. 37 patients were evaluable for response,
`31 (84%) achieved CRc. This data compares favorably
`to the CRc of 54% with single agent gilteritinib in the
`ADMIRAL study; suggesting gilteritinib plus veneto-
`clax might be better option for R/R FLT3-mutated AML,
`while longer follow-up with OS data is awaited [56]. A
`Phase Ib/II trial explored the combination of quizartinib
`(Quiz) with decitabine (10  days) ± venetoclax mostly in
`patients with R/R AML. CRc of 90% was achieved in the
`DEC10 + VEN + Quiz cohort, and CRc rate of 40% was
`achieved in DEC10 + quiz cohort. In addition, CyTOF
`(single-cell mass cytometry) analysis could be used to
`select patients with the best response based on pre- and
`on-therapy apoptotic and signaling pathway profiles [57].
`
`Targeting TP53 mutation
`The TP53 gene, located on chromosome 17p13.1, is com-
`monly mutated in tumors making it one of the most
`widely mutated genes in human malignancies. TP53
`mutations are detected in 5% to 20% of patients with
`newly diagnosed AML and MDS, with higher incidences
`in older patients and in those with secondary AML or
`therapy-related myeloid neoplasms. TP53 mutation is
`
`enriched in patients with complex karyotype and mon-
`osomal karyotypes and also in patients with relapse or
`refractory disease. TP53 mutation has been associated
`with a poor prognosis in both AML and MDS [58, 59].
`A recent study analyzed 3,324 patients with MDS for
`TP53 mutations and allelic imbalances, and deline-
`ated two subsets of patients with distinct phenotypes
`and outcomes. One-third of TP53-mutated patients had
`monoallelic mutations whereas two-thirds had multi-
`ple hits consistent with biallelic targeting. Established
`associations with complex karyotype, few co-occurring
`mutations, high-risk presentation and poor outcomes
`were specific to multi-hit patients only. The TP53 multi-
`hit state predicted a high risk of death and leukemic
`transformation independently of the revised interna-
`tional prognostic scoring system (IPSS-R). Importantly,
`monoallelic patients did not differ from

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