Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Apr 1.
Published in final edited form as: Curr Hematol Malig Rep. 2021 Mar 18;16(2):192–206. doi: 10.1007/s11899-021-00621-9

Novel Targeted Therapeutics in Acute Myeloid Leukemia: An Embarrassment of Riches

Nicole R Grieselhuber 1, Alice S Mims 1
PMCID: PMC8515252  NIHMSID: NIHMS1701097  PMID: 33738705

Abstract

Purpose of review:

Acute myeloid leukemia (AML) is an aggressive malignancy of the bone marrow that has a poor prognosis with traditional cytotoxic chemotherapy, especially in elderly patients. In recent years, small molecule inhibitors targeting AML associated IDH1, IDH2 and FLT3 mutations have been FDA approved. However, the majority of AML cases do not have a targetable mutation. A variety of novel agents targeting both previously untargetable mutations and general pathways in AML are currently being investigated. Herein, we review selected new targeted therapies currently in early phase clinical investigation in AML.

Recent Findings:

The DOT1L inhibitor pinometostat in KMT2A rearranged AML, the menin inhibitors KO-539 and SYNDX-5613 in KMT2Ar and NPM1 mutated AML and the mutant TP53 inhibitor APR-246 are examples of novel agents targeting specific mutations in AML. In addition, BET inhibitors, polo-like kinase inhibitors and MDM2 inhibitors are promising new drug classes for AML which do not depend on the presence of a particular mutation.

Summary:

AML remains in incurable disease for many patients but advances in genomics, epigenetics and drug discovery have led to the development of many potential novel therapeutic agents, many of which are being investigated in on-going clinical trials. Additional studies will be necessary to determine how best to incorporate these novel agents into routine clinical treatment of AML.

Keywords: Acute Myeloid Leukemia, Targeted Therapy, DOT1L, MDM2 inhibitors, bromodomain inhibitors, polo-like kinase inhibitors, menin inhibitors

Introduction

Acute myeloid leukemia (AML) is a heterogeneous malignancy of hematopoietic stem and progenitor cells that has a poor prognosis with standard cytotoxic chemotherapy (1). In the last decade, significant progress has been made in defining the cytogenetic, molecular and epigenetic alterations underlying leukemogenesis (24). Response to cytotoxic chemotherapy and therefore prognosis is highly dependent upon cytogenetic and mutational aberrations present. The identification of AML associated mutations has led to the development of new classes of targeted inhibitors including the mutant IDH inhibitors enasidenib (5, 6) and ivosidenib (7, 8) and the FLT3 inhibitors midostaurin (9) and gilteritinib (10). However, only a minority of AML patients have targetable mutations. Targeted inhibitors that block pathways broadly involved in AML and that are not dependent upon specific mutations are another strategy; two such drugs have recently been approved. The BCL2 inhibitor venetoclax has been approved in combination with hypomethylating agents or low dose cytarabine (LDAC) for older (≥75 years) and/or unfit AML patients.(11, 12). The smoothened inhibitor glasdegib is similarly approved to be given in combination with LDAC (13). While these new agents have improved outcomes in AML, none are considered curative. There remains an unmet need for additional novel targeted therapeutics, particularly in patients with adverse risk disease. In this article, we will review selected targeted inhibitor classes currently under development for the treatment of AML.

BET Inhibitors

The epigenetic reader bromodomain and extraterminal domain (BET) protein BRD4 was initially identified as an essential gene for the survival of MLL-AF9 / NRAS driven murine AML cells (14) and has been shown to have a crucial role in maintaining survival in human AML leukemia stem cells (LSC) (15). BRD4, especially the short isoform, is over-expressed in human AML and high risk myelodysplastic syndrome (MDS) (16, 17). Collectively, evidence suggests that BRD4 serves to maintain core transcriptional programs in LSCs (18). It should be noted that little is known about the role of the other ubiquitously expressed BET family members BRD2 and BRD3 in leukemogenesis.

Pharmacologic inhibitors of BRD4 have been developed which function via displacing BRD4 as well as BRD2 and BRD3 from acetylated lysines on histones and transcription factors (19, 20). BET inhibitor treatment of AML cells results in differentiation and loss of LSC associated gene expression signatures (14, 19, 21). In addition to their initially reported activity in mixed-lineage leukemia 1 / lysine-specific methyltransferase 2A (MLL1/KMT2A) rearranged (KMT2Ar) AML, these drugs have been shown to have pre-clinical activity against multiple sub-types of AML, including NPM1 mutated (21), IDH2 mutated (22), ASXL1 mutated (23), t(8;21) positive core binding factor (CBF) AML (24) and JAK2 mutated secondary AML (25). These pre-clinical data suggest that BET inhibitors may have broad activity in AML that is not limited specific mutations or cytogenetic abnormalities.

Multiple BET inhibitors are currently in pre-clinical and early clinical development, with significant differences in selectivity, half-life and other pharmacologic properties. Early BET inhibitors were panselective, with relatively indiscriminant binding between both different BET proteins and BD1/BD2 domains within a given BET protein (26). Subsequent work has shown that BD1 may be more critical for continued chromatin binding, while the role of BD2 may be limited to initiating binding and interactions with transcription factors (27). Additionally, BD2 seems to have more relevance for inflammatory responses while transcriptional networks in AML may be BD1 dependent (28). There still is much to be learned regarding the relative roles of different BET proteins and specific bromodomains in AML, and it remains unclear which bromodomain should be pharmacologically targeted for optimal responses.

To date, the final results of only one clinical trial of BET inhibitors in AML has been fully reported (29). In this study OTX015 / MK-8628 was given in phase 1 dose escalation study in relapsed and refractory (r/r) acute leukemia patients. On the basis of dose limiting toxicities (DLTs), including gastrointestinal (GI) and thrombocytopenia, when given continuously in a separate study in lymphoma and multiple myeloma (MM) patients (30), OTX015 was given for 14 days followed by 7 days off drug. Dose escalation was hampered by poor tolerability secondary to GI toxicities, fatigue and cytopenias, particularly at doses higher than 80 mg daily. Responses included 2 complete remissions (CR) and 1 CR with incomplete count recovery (CRi) with no apparent relationship between dose level and responses. Duration of response was short, with all 3 responding patients relapsing within 5 months or less. OTX015 is no longer being clinically developed; however multiple trials of second and third generation BET inhibitors, both as single agents and in combination with hypomethylating agents or venetoclax are currently on-going as summarized in Table 1.

Table 1.

Current Clinical Trials with BET Inhibitors

Drug Selectivity Sponsor NCT ID Phase Disease Combination Status Reference
ABBV-075 / Mivebresib Pan-BET AbbVie NCT02391480 1 AML Venetoclax Completed 144
ABBV-075 / Mivebresib Pan-BET AbbVie NCT04480086 1 MF Ruxolitinib or navitoclax Recruiting 145
ABBV-744 BD2 AbbVie NCT03360006 1 AML None Recruiting
ABBV-744 BD2 AbbVie NCT04454658 1 MF Ruxolitinib or navitoclax Recruiting 145
AZD5153 Bivalent 107 NCT03013998 1/2 AML Venetoclax Recruiting
CPI0610 Pan-BET Constellation Pharmaceuticals NCT02158858 2 MF Ruxolitinib Recruiting 146148
CPI0610 Pan-BET Constellation Pharmaceuticals NCT04603495 3 MF Ruxolitinib Recruiting 149
FT-1101 Pan-BET Forma Therapeutics NCT02543879 1 AML, MDS, NHL None Completed 150
GSK525762 / Molibresib Pan-BET GlaxoSmithKline NCT01943851 1 AML, NHL, MM None Completed 151
INCB054329 Pan-BET Incyte Corporation NCT02431260 1 AML, MDS/MPN, MF None Terminated
INCB057643 Pan-BET Incyte Corporation NCT04279847 1 MF None Recruiting 152
PLX2853 Modestly BD2 Plexxikon NCT03787498 1 AML, MDS None Recruiting 153
PLX51107 Pan-BET MDACC/NCI NCT04022785 1 AML, MDS Azacitdine Recruiting
PLX51107 Pan-BET Plexxikon NCT02683395 1 AML, MDS, lymphoma None Terminated 154
RO6870810 / TEN-10 Pan-BET Roche NCT02308761 1 AML, MDS None Completed 155

Abbreviations: AML, acute myeloid leukemia; BET, bromodomain extraterminal domain; MDS, myelodysplastic syndrome; MF, myelofibrosis; MM, multiple myeloma; MPN, myeloproliferative neoplasm; NHL, non-hodgkin lymphoma

DOT1L inhibitors

Approximately 5–10% of AML cases harbor 11q23 chromosomal rearrangements involving the MLL1/KMT2A histone methyltransferase (2, 3). KMT2Ar are disproportionately common in pediatric and young adult patients (31) and in patients with prior exposure to topoisomerase II inhibitors (32). These patients have an aggressive course with inferior responses to chemotherapy (33). KMT2A fusion proteins inappropriately recruit disruptor of telomeric signaling −1 (DOT1L), a H3K79 methyltransferase, to KMT2A target genes, in particular the HOXA cluster and MEIS1, resulting in deposition of activating histone methylation marks at these promoters and ultimately overexpression (34). Genetic screens demonstrated that KMT2A fusions require DOT1L activity for leukemic transformation (35), making DOT1L an attractive pharmacologic target. Subsequently, EPZ004777, a potent and selective inhibitor of DOT1L, was developed (36). EPZ004777 has little effect on leukemia cells without KMT2Ar, but decreases proliferation and induces apoptosis and differentiation in KMT2Ar cells. Importantly, given that greater than 60 types of 11q23 translocation partners have been described to date (37), EPZ004777 has activity against multiple KMT2A fusion proteins (33, 36). However, EPZ004777 has unsuitable pharmacokinetic (PK) properties for in vivo use, prompting the development of an optimized compound, pinometostat (EPZ-5676), for clinical development (38). Pinometostat is highly potent DOT1L inhibitor with >37,000 fold selectivity over other protein methyltransferases. In subcutaneous (SQ) MV4–11 xenograft models, pinometostat treatment resulted in tumor regression (38). In pre-clinical models, continuous drug exposure was required for efficacy, with decreased responses seen with intermittent dosing. Therefore, pinometostat was given as a continuous IV infusion for 21 days of a 28 day cycle in the initial early phase clinical studies. In a multi-center dose escalation study in 51 r/r acute leukemia patients, of which 37 were KMT2Ar, pinometostat was well tolerated, with the most common adverse events being fatigue, nausea, febrile neutropenia and electrolyte abnormalities (39). Serious (>grade 3) treatment related adverse events (AEs) included leukocytosis possibly related to differentiation, anemia, prolonged QTc, cardiac failure and liver abnormalities. Two patients had objective CRs and two patients had clearance of leukemia cutis lesions without regression of bone marrow disease. Additionally, seven patients had leukocytosis with morphologic changes on bone marrow examination consistent with differentiation. A similar safety profile was seen in a dose escalation study in pediatric KMT2Ar acute leukemia patients (40), with the exception of cardiac toxicity, which was only observed in adult patients. In contrast to the initial study in adult patients, pinometostat was dosed continuously until progression or unacceptable toxicity. No objective responses were obtained, but 7/18 patients had transient decreases in blood or bone marrow blast percentage.

On the basis of these results in heavily pre-treated r/r patients, pinometostat is currently being investigated in two phase 1b/2 studies in newly diagnosed KMT2Ar AML patients. In NCT03724084, pinometostat is given in combination with daunorubicin and cytarabine-based induction chemotherapy in patients ages 14 or older who are candidates for intensive chemotherapy. In NCT03701295, either 54 mg or 90 mg/m2/day pinometostat is given continuously in combination with azacitidine either in newly diagnosed KMT2Ar AML patients ineligible for induction chemotherapy or in r/r KMT2Ar AML. Recruitment for these trials is on-going and results have not yet been reported.

Polo-like kinase inhibitors

Polo-like kinases are a family of serine/threonine kinases with roles in cell cycle regulation and characterized by the presence of a conserved C-terminal polo-box domain that mediates protein-protein interactions with substrates (41). The best studied family member, polo-like kinase 1 (PLK1) is required for cells to progress from G2 to M phase in the cell cycle and is characteristically overexpressed in rapidly dividing cells including during fetal growth and in various malignancies (42) including AML (43). Additionally, PLK1 interacts with the PI3K/AKT/MTOR pathway, particularly in leukemia cells (44). There is little to no PLK1 expression in normal, adult tissues (43) including CD34+ progenitors and PBMCs (43, 45). Genetic or pharmacologic inhibition of PLK1 in leukemia cells results in cell cycle arrest in pro-metaphase leading to apoptosis and decreased colony formation (43, 46), with little effect on normal CD34+ cells. Importantly, the effects of PLK1 depletion or inhibition do not appear dependent upon the presence of TP53 (42). Recent pre-clinical data suggests that complex karyotype AML (47), aneuploid AML (48) and secondary AML (49) may be particularly dependent upon PLK1 and therefore especially sensitive to PLK1 inhibition.

The first PLK1 inhibitor studied clinically, BI-2536, (46) was investigated in elderly r/r AML patients in a multi-center phase 1 dose escalation study (50). The toxicity profile was typical of AML patients, with the majority of adverse events being hematologic and infectious in nature. Encouragingly, 2 CRs and 3 partial responses (PRs) were observed in a heavily pretreated population. Of patients whose response was initially classified as a PR, all three eventually achieved a CR with additional cycles of treatment. One of these patients maintained a CR for 21 months. Similar to pre-clinical data, bone marrow cells 24 hours after BI-2536 infusion exhibited mitotic arrest and apoptosis. Despite these encouraging results, development of BI-2536 was discontinued due to a multi-compartmental PK behavior thought to be unfavorable for clinical use.

Several PLK1 inhibitors are currently being studied in early phase clinical trials (Table 2). Of these, volasertib, a derivative of BI-2536, has the most data. Volasertib is potent competitive inhibitor of PLK1 with a sub-nanomolar IC50, little activity on unrelated kinases (51) and improved pharmacologic properties compared to pre-clinical PLK1 inhibitors (52). It has additional inhibitor activity on BRD4 (53) though the relevance of this in vivo is unclear. In the first-in-human phase 1 dose escalation study of volasertib given as a 1 hour IV infusion every 3 weeks in advanced or metastatic solid tumors, volasertib was well tolerated, with the main AEs being cytopenias and nausea (54). Neutropenia and thrombocytopenia were dose limiting but reversible. The maximum tolerated dose (MTD) was found to be 400 mg. PRs were seen in 3 of 65 patients and stable disease (SD) was observed in an additional 26 subjects. A similar toxicity profile was seen in two additional phase 1 studies in advanced solid tumors (55, 56) and in r/r AML patients receiving either volasertib monotherapy or volasertib in combination with LDAC (57). The MTD in combination with LDAC was determined to be 350 mg. Subsequently, volasertib was investigated in a phase 2 study that randomized 89 older AML patients unsuitable for induction chemotherapy to LDAC (20 mg SQ twice daily on days 1–10) with or without volasertib (350 mg on days 1 and 15) (58). The composite CR/CRi rate was significantly higher in patients receiving LDAC + volasertib (12/42, 31%) compared to LDAC alone (6/45, 13.3%). Combination arm patients also had longer event-free survival (EFS) (median 5.6 vs 2.3 months) and overall survival (OS) (median 8.0 vs 5.2 months). Responses were not correlated with cytogenetic risk classification. Unfortunately, the POLO-AML2 phase 3 study of LDAC + volasertib versus LDAC + placebo did not meet its primary endpoint of improved CR/CRi (59). The combination arm did have a higher CR/CRi rate (25.2 vs 16.8%) which was not statistically significant. The initial analysis showed an increased incidence of fatal infections in the volasertib arm, which was related to total dose density. Subsequently, a phase 1 study of volasertib in combination with decitabine was conducted in 13 elderly AML patients (60). The MTD of volasertib when given in combination with decitabine was 400 mg on days 1 and 5. Two patients had DLTs, including grade 3 asthenia and grade 3 mucositis, at the 400 mg dose level. Additionally, treatment related grade 5 anemia occurred in one patient; this was not considered a DLT due to the study protocol defining only non-hematologic toxicities as DLTs. Three objective responses, 2 CRi and 1 PR, were reported. An additional study of volasertib in combination with azacitidine in MDS (NCT02721875) has been terminated but final results have not yet been reported.

Table 2.

Current Clinical Trials with PLK Inhibitors

Drug Selectivity Sponsor NCT ID Phase Disease Combination Status Reference
BI-2536 Pan PLK Boehringer Ingelheim NCT00701766 1 AML None Completed 50
CFI-400945 PLK4 University Health Network Toronto, Canada NCT03187288 1 AML, MDS None Recruiting 156
CFI-400945 PLK4 Treadwell Therapeutics, Inc NCT04730258 1/2 AML, MDS, CMML Azacitidine or decitabine Not yet recruiting 157
CYC140 PLK1 Cyclacel Pharmaceuticals NCT03884329 1 AML, ALL, MDS, CML, CLL None Recruiting
Onvansertib PLK1 Cardiff Oncology NCT03303339 1/2 AML LDAC or decitabine Active, not recruiting 66, 67
Rigosertib PLK/RAF/PI3K inhibitor Onconova Therapuetics NCT01926587 1/2 AML, MDS, CMML Azacitidine Active, not recruiting 158
Rigosertib PLK/RAF/PI3K inhibitor Onconova Therapuetics NCT00854646 1 AML, MDS, CMML, CML, CLL None Completed 159
Rigosertib PLK/RAF/PI3K inhibitor Onconova Therapuetics NCT01241500 3 MDS None Completed 160
Rigosertib PLK/RAF/PI3K inhibitor Onconova Therapuetics NCT02562443 3 MDS None Active, not recruiting
Volasertib Pan PLK Boehringer Ingelheim NCT00804856 1 AML LDAC Active, not recruiting 57
Volasertib Pan PLK Boehringer Ingelheim NCT00804856 2 AML LDAC Active, not recruiting 58
Volasertib Pan PLK Boehringer Ingelheim NCT01721876 3 AML LDAC Active, not recruiting 59
Volasertib Pan PLK Boehringer Ingelheim NCT02003573 1 AML Decitabine Terminated 60
Volasertib Pan PLK Boehringer Ingelheim NCT02721875 1 MDS Azacitidine Terminated

Abbreviations: AML, acute myeloid leukemia; ALL, acute lymphoblastic leukemia; CLL, chronic lymphocytic leukemia; CML, chronic myeloid leukemia; CMML, chronic myelomonocytic leukemia; LDAC, low-dose cytarabine; MDS, myelodysplastic syndrome; PLK, polo-like kinase

At this time, further development of volasertib in AML has been discontinued. The toxicities and limited clinical efficacy of volasertib was thought to be potentially related to off-target effects on other polo-like kinase family members PLK2 and PLK3. Less is known about the physiologic role of PLK2 and PLK3 in hematopoiesis. Some studies suggest that PLK2 may have tumor suppressor activity in hematologic malignancies including AML (61) and its suppression may increase senescence and apoptosis in normal marrow progenitors (62). Onvansertib/NMS-P37/PCM-075 is a potent reversible, competitive orally bioavailable selective PLK1 inhibitor that has limited off target activity on PLK2 and PLK3 at concentrations up to 10 μM (63, 64). Additionally, onvansertib has a shorter half-life than volasertib, which may also contribute to decreased toxicity.

In a phase 1 dose escalation study of onvansertib given days 1–5 of 21 day cycles to advanced/metastatic solid tumor patients, no DLTs were observed up to 24 mg/m2/day (65). Drug-related grade 4 neutropenia and thrombocytopenia were seen at higher doses. SD was the best response and was achieved in 5 of 16 evaluable patients. KRAS mutations appeared to be a biomarker of response in metastatic colon cancer and onvansertib has been granted Fast Track designation by the United States Food and Drug Administration (FDA) for further development in this patient population. In AML, onvansertib was investigated in a phase 1b dose escalation study in r/r patients in combination with LDAC or decitabine, with a starting dose of 12 mg/m2 on days 1–5 of a 28 cycle (66). Overall, ovanasertib was well tolerated, with the most common adverse events being hematologic toxicities. Two DLTs, grade 3 mucositis and grade 4 rash, occurred at 90 mg/m2 in the decitabine arm. The MTD for onvansertib was therefore established as 60 mg/m2. CR or CRi was reported in 5 patients (5/23, 24%) on the decitabine arm and 1 patient (1/15, 7%) on the LDAC arm, with a median remission duration of 5.5 months. Notably, while the study was not powered to detect associations between different mutational subgroups, 4 of the 6 patients who attained CR or CRi had spliceosome mutations. An update of the phase 2 expansion study of 60 mg/m2onvansertib in combination with decitabine was recently reported at the 2020 American Society of Hematology meeting (67). The toxicities were similar to those seen in the phase 1b study. The combined CR/CRi rate was 17%, with half of responders having a spliceosome mutation.

Rigosertib is a small molecule with inhibitory activity on polo-like kinases, RAF and PI3K (68) that has been investigated in AML and MDS. It is not yet clear to what extent its activity its activity depends upon PLK versus RAS pathway inhibition. Due to space constraints, we will not discuss rigosertib further in this review.

Mutant TP53 inhibitors

TP53 is a tumor suppressor protein with roles in DNA damage response, cell cycle arrest, apoptosis and senescence (69). Approximately 5–10 percent of AML cases have TP53 mutations (2, 3), with an increased prevalence in therapy related-AML (70), secondary AML (71) and complex-karyotype AML (72). TP53 mutated AML has a poor prognosis, with inferior responses to traditional cytotoxic chemotherapy (7375). Interestingly, AML with subclonalTP53 mutations has a similarly poor prognosis compared to cases in which the dominant clone is TP53 mutated (76). Responses may be more favorable with decitabine-based therapy (77), though this may be altered by the effect of concurrent karyotype abnormalities on responses in this patient population (78, 79).

A novel therapeutic strategy is to target mutated TP53, a protein which had previously been considered undruggable. APR-246, a PRIMA-1 analog, binds covalently to TP53 and induces a conformational shift in mutated TP53 to a wildtype-like structure which is able to bind specific DNA sequences and activate target gene transcription, thereby ultimately inducing apoptosis (80, 81). In addition, APR-246 has TP53-independent activity that leads to increased oxidative stress and cell death (82), providing a plausible mechanism for activity in TP53 wildtype cells and in cells without detectable TP53 expression. In preclinical models of AML, APR-246 has synergistic effects with azacitidine (83).

In a first-in-human phase 1/2 dose escalation study in patients with refractory hematologic malignancies and advanced prostate cancer, APR-246 was given as a 2 hour IV infusion for 4 consecutive days, followed by a 17 day safety monitoring period (84). Retreatment was only allowed if disease burden was judged to have decreased by 25% or greater after initial treatment. APR-246 was well tolerated, with the most common AEs being fatigue, dizziness, headache, confusion and sensory disturbances. Neurologic side effects were infusion-related and resolved after the end of infusion. The MTD was determined to be 60 mg/kg. One AML patient had a reduction in bone marrow blasts from 46% to 26% and one non-Hodgkin Lymphoma patient had a minor response on computed tomography; both of these individuals had TP53 mutated disease. An extension of this study in 8 AML and 2 chronic lymphocytic leukemia (CLL) patients sought to improve safety by extending the infusion time to 6 hours and additionally allowed for retreatment in 21 day cycles (85). Using this strategy, the MTD was increased to 105 mg/kg. Two AML patients met response criteria. One patient achieved a CR, though interpretation of this observation is complicated by use of hydrea and sorafenib within 4 weeks of treatment. TP53 mutational status was not determined for this patient. A second TP53 mutated AML patient had a reduction in peripheral blood blast count >25% at 21 days but was taken off study and did not have additional response assessment or treatment due to AEs thought unrelated to the study drug. A third patient with TP53 mutated CLL had a transient >25% decrease in total lymphocyte count and lymph node size after 1 cycle of treatment but progressed during cycle 2. Notably in both of these studies, clinical activity in hematologic malignancies was concentrated in TP53 mutated patients

Based on these results, APR-246, now known as eprenetapopt, was then studied in combination with azacitidine in two early phase studies in TP53 mutated AML and MDS patients. Sallman et al reported the results of a phase 1b/2 of APR-246 alone or in combination with azacitidine in TP53 mutated myeloid malignancies including 40 MDS, 11 AML and 4 chronic myelomonocytic leukemia patients (86). In the phase 1b study, dose escalation up to 100 mg/kg eprenetapopt given IV on days 1–4 was well tolerated without DLTs either as monotherapy or in combination with azacitidine given on days 4–10. The RP2D of 100 mg/kg was determined to be equivalent to a fixed dose of 4500 mg daily on the basis of PK data; this dosing was used in the phase 2 expansion. The AEs were similar to those associated with each drug as a single agent and included cytopenias, febrile neutropenia, nausea/vomiting, diarrhea and transient neurologic toxicities during eprenetapopt infusion. Grade 3/4 toxicities were limited to cytopenias; there were no reported serious neurologic toxicities. Of 45 evaluable patients, 36 achieved CR or marrow CR, including 6 of 8 evaluable AML patients. The median duration of CR was 7.3 months; this was not significantly different between MDS and AML patients. Notably, 19 patients discontinued study treatment in order to undergo allogeneic stem cell transplant.

A Groupe Francophone des Myélodysplasies phase 2 study of APR-246 4500 mg IV days 1–4 followed by azacitidine given days 4–10 of a 28 day cycle was published concurrently (87). Fifty-two patients, including 34 MDS and 18 AML patients, were evaluable for response. The overall response rate (ORR) was 52% including 37% CR. The reported CR rate in MDS patients was 53%. AML patients had lower CR/CRirates, with a trend towards decreased efficacy in patients with >30% marrow blasts (14% vs 45% for patients with 20–30% marrow blasts). Adverse events were similar to those reported in the phase 1b/2 study (86). Two reversible grade 3 neurologic toxicities were observed in patients with baseline lower renal function. In both of these studies, TP53 mutations VAF clearance was associated with CR. There was no clear relationship between co-occurring mutations and response.

On the basis of these results, the FDA granted APR-246 and azacitidine breakthrough designation for TP53 mutated MDS in April 2020. However, the randomized phase 3 study of APR-246 and azacitidine compared to azacitidine (NCT03745716) was recently reported to not meet the primary endpoint of CR. However, further analysis and full report of the study is still pending at the publication of this manuscript. APR-246 is also being investigated as maintenance therapy in combination with azacitidine following alloSCT in TP53 mutated AML or MDS (NCT03931291) and in combination with azacitidine/venetoclax for newly diagnosed TP53 mutated myeloid malignancies (NCT04214860). In addition, Aprea Therapeutics is currently developing an orally bioavailable APR-246 derivative, APR-548. A phase 1 study of APR-548 in combination with azacitidine in TP53 mutated MDS is planned but not yet recruiting (NCT04638309).

MDM2 Inhibitors

TP53 activity is inhibited by binding its negative regulators MDM2 and MDM4, which target it for degradation via ubiquitination. MDM2 is overexpressed in AML so even cells without TP53 mutation or deletion may have low functional TP53 activity. Small molecule inhibitors of the MDM2/p53 interaction reduce p53 destruction leading to increased cellular p53 levels and ultimately apoptosis (88). The first inhibitor of this class, nutlin-3a, was not studied clinically due to poor bioavailability and toxicity in preclinical models. Subsequently, two nutlin-3a analogues, RG7112 and RG7388, were developed. RG7112 was studied in a phase 1 trial in r/r acute and chronic leukemia patients, with modest efficacy (89) including 3 CR/CRi out of 30 AML patients. However, its further development was also limited by GI toxicity (90) and unfavorable PK findings.

RG7388/idasanutlin is an orally bioavailable MDM2 inhibitor with increased potency and selectivity compared to both RG7112 and nutlin-3a (91). In a phase 1 dose escalation study of r/r AML, idasanutlin was given either as a single agent on days 1–5 of a 28 cycle or in combination with cytarabine 1 g/m2 IV on days 1–6 of a 28 day cycle (92). Enrollment was not limited to TP53 wild-type AML. Myelosuppression and diarrhea were identified as DLTs, with both occurring at the 800 mg twice daily dose level. Two different formulations of idasanutlin were studied, with different recommended phase 2 dosing determined for each: 600 mg twice daily for the initial formation and 300 mg twice daily (BID) for the optimized formulation used in an expansion study. Seven of 37 evaluable patients (19%) receiving monotherapy achieved a CR or CRi, while 21 of 59 (36%) evaluable combination treatment patients obtained a CR or CRi. The median duration of response was 7.7 months with monotherapy and 8.5 months with combination therapy. Six patients proceeded to alloSCT. Notably, only 1 of 25 patients with TP53 mutations attained CR, compared to 22 of 71 TP53 wild-type patients. Responses were additionally correlated with MDM2 expression in AML blasts (92, 93). Idasanutlin was additionally studied at a lower dose in polycythemia vera and essential thombocythemia patients refractory to standard of care treatments (94), where it was well tolerated. GI toxicities including nausea, vomiting and diarrhea were the most common AEs but these were generally low grade. Of 16 enrolled patients, responses were observed in 9, including 5 CRs and 4 PRs. Unfortunately, despite these promising results, the phase III MIRROS study of idasanutlin in combination with cytarabine (NCT02545283) in r/r AML (95) was terminated early when it failed to meet its primary endpoint of improved overall survival compared to cytarabine alone. It is important to note that the bioavailability and PKs of idasanutlin are heavily influenced by formulation (9698). It is therefore difficult to directly compare results from initial studies to later trials using an optimized formulation. Currently idasanutlin is being investigated in combination with venetoclax in r/r AML (NCT02670044 and NCT04029688) and in combination with cytarabine and daunorubicin based induction chemotherapy in newly diagnosed AML (NCT03850535). Preliminary results of the phase 1 study investigated idasanutlin and venetoclax in elderly r/r AML and secondary AML were reported at the 2019 American Society of Hematology annual meeting (99). Adverse events were consistent with known toxicities of both drugs as single agent therapy. The RP2D was determined to be 600 mg venetoclax given continuously in combination with either 150 mg or 200 mg of idasanutlin on days 1–5 of 28 day cycles. Across all dose levels, CR or CRi were recorded in 11 of 49 patients.

The MDM2 inhibitor AMG-232 is structurally distinct and interacts with the glycine shelf region of the TP53 binding pocket on the surface MDM2 (100), a property not found in nutlin derivatives. Like idasanutlin, it is orally bioavailable. In a phase 1 dose-escalation study in advanced solid malignancies and MM, AMG-232 was well tolerated, with the most common adverse events being GI toxicities and anemia (101). No objective response were reported. In a second study, AMG-232 was given alone or in combination with the mitogen-activated protein kinase (MEK) inhibitor trametinib in r/r AML (102). Of note compared to the first-in-man study in solid tumor patients, this study had increased dose frequency with AMG-232 being given for 7 days every 2 weeks rather than 7 days every 3 weeks. Despite the increased dosing, the toxicity profile was similar. As expected, AML patients had a higher incidence of grade 3 and 4 cytopenias. Dose escalation in the monotherapy arm was halted due to GI toxicities. However, the protocol did not allow prophylactic medications to manage nausea or diarrhea and the authors note that tolerability may be improved with prophylactic anti-emetics. Responses included 4 morphological leukemia free states (MLFS) in the monotherapy arm and 1 CR and 1 PR in the combination arm out of a total 30 evaluable patients. As expected based on the mechanism of action, no responses were seen in patients with TP53 mutations. Most responses were short lived. However, the patient who attained a CR maintained this response for over 550 days. Trametinib was discontinued after cycle 2 due to trametinib-related ocular toxicity and AMG-232 was continued as a single agent until disease progression 650 days after initiating study treatment. AMG-232 is currently being investigated in combination with decitabine (NCT03041688 and NCT04113616), LDAC (NCT04113616) and with induction chemotherapy (NCT04190550) in AML

One limitation of MDM2 inhibitors is that most compounds have limited effects on other MDM family members such as MDMX, which may be a source of resistance. AML cells have high MDMX expression (102); this may present difficulties for the development of MDM2 inhibitors. ALRN-6924 is a stapled peptide with dual inhibition of both MDM2 and MDM4 (103). Notably it is the first stapled peptide to be given in man. In a first-in-human phase 1 study in advanced solid tumorand lymphoma patients, ALRN-6924 was generally well tolerated with the most common AEs being cytopenias, GI toxicity, liver function test abnormalities and infusion reactions, all of which were reversible. ALRN-6924 has been investigated in a phase 1 study in r/r AML and MDS, both as a single agent and in combination with cytarabine. Final results have not yet been reported but in preliminary results presented in abstract form (104), no DLTs had been observed and the MTD had not yet been reached. Marrow CR was reported in 2 MDS patients.

Menin Inhibitors

Several genetic subgroups of AML are characterized by overexpression of the homeobox (HOX) A family of transcription factors, including KMT2Ar (105), KMT2A partial tandem duplication (PTD) mutated (106) and NPM1 mutated AMLs (107109). As discussed above, KMT2Ar AML is a particularly aggressive subtype of AML. KMT2A-PTD occurs in 10% of adult AML cases (110) and is also associated with poor prognosis (111). Mutations in the nuclear chaperone protein nucleophosmin (NPM1), which result in mis-localization of NPM1 to the cytoplasm (112), occur in 25–30% percent of adult AML cases (24) and up to 10% of pediatric and young adult AML (113).

HOX genes serve as master regulators of hematopoiesis, controlling both self-renewal in normal hematopoietic stem cells and differentiation to committed myeloid progenitors (114, 115). The understanding of how KMT2A fusions and KMT2A-PTD proteins deregulate HOX gene expression has been elegantly investigated by multiple groups over the last twenty years. In normal hematopoiesis, KMT2A is a crucial transcriptional regulator of HOX genes (116). Similarly, KMT2A fusion and KMT2A-PTD proteins bind to the promoters of HOXA genes and activate transcription (105, 117, 118) via recruitment of a complex of chromatin remodeling proteins comprising the adaptor protein menin (119121) in addition to DOT1L (34, 35) and BET proteins (14) as discussed above. The interaction of menin with both wild type KMT2A (119) and KMT2A fusion proteins (120) is necessary for HOX gene expression (121, 122). The mechanism by which cytoplasmic NPM1 induces HOX expression is less clear, but it seems to be a direct consequence of the NPM1 mutation since expression of a mutated NPM1 allele in mice also results in increased expression of HOXA genes (123125). In addition, genetic or pharmacologic abrogation of cytoplasmic NPM1 reduced HOX gene expression and produced cell cycle arrest and differentiation in NPM1 mutated cell lines, patient derived xenografts and genetically engineered human HSCs (126, 127). Like KMT2Ar and KMT2A-PTD, AML NPM1 mutated leukemia cell lines require wildtype KMT2A and menin to maintain HOX expression (128).

The role of menin in maintaining self-renewal in multiple subtypes of AML made it an attractive pharmacologic target. However, disrupting large protein-protein interactions pharmacologically was technically challenging and for many years, this was thought to be undruggable. Recent developments have challenged those assumptions. Detailed structural studies that accurately modeled the interaction between menin and KMT2A, especially the KMT2A binding pocket on menin were a pre-requisite to successful drug development (129). The first pre-clinical small molecule inhibitor of menin / KMT2A1 interaction was MI2–2, a thienopyrimidine compound developed at the University of Michigan (130). Unlike DOT1L inhibitors, which inhibit the H3K79 methyltransferase enzymatic activity of the KMT2A fusion protein complex, menin inhibitors disrupt formation of the complex (131). MI2–2 and related compounds had encouraging pre-clinical in vitro activity, inducing cell cycle arrest, apoptosis, differentiation and reduction in KMT2A fusion protein target gene expression, including HOX genes (130, 132). Importantly, activity was independent of the KMT2A fusion partner (132). However, poor pharmacokinetic properties limited in vivo studies. Therefore, a series of optimized derivative compounds with improved potency, reduced off-target effects and oral bioavailability was developed (133). MI-3454, the most recently reported of this family of compounds, induced complete remissions in both KMT2Ar and NPM1 mutated patient derived xenograft (PDX) mice with little effect on normal hematopoiesis (134). Similarly, KO-539, a structurally related analog of MI-3454, induced in vivo differentiation, complete remissions and long lasting survival in KMT2Ar cell line xenografts (135, 136) and NPM1 mutated PDX models (137).

KO-539 is currently being investigated in phase 1/2a study in r/r AML (NCT04067336) and has been granted orphan drug designation by the FDA. Notably, the phase 1 study does not include a requirement for NPM1 mutations or KMT2Ar. In preliminary results reported at the 2020 ASH annual meeting (138), 12 patients had been enrolled at doses ranging from 50–400 mg once daily dosed continuously. Importantly, no dose limiting toxicities have been identified and no patients discontinued treatment due to adverse events. The most common toxicities included nausea, diarrhea and rash, all of which were grade 1 or 2. KO-539 pharmacokinetics were not altered by co-administration of CYP3A4 inhibitors, such as azole antifungals. Of 8 evaluated patients, 6 had evidence of clinical activity. These included a NPM1, DNMT3A mutated AML that attained MRD negative complete remission, a MRD positive CR in a SETD, RUNX1 mutated AML and a MLFS in a NPM1 mutated, FLT3-ITD AML. Additionally a patient with KMT2Ar AML had a decrease in hydrea requirements when treated with the lowest dose, 50 mg daily. Other reported responses were decreasing peripheral blasts and SD in one patient each. A CR occurring in a patient without NPM1 mutations or KMT2A rearrangement raises the possibility that KO-539 may have broader applications in the treatment of AML. This is in line with pre-clinical data showing menin’s role in maintenance of transcriptional programs in other subtypes of AML, including MN1-ETV6 translocations (139), FLT3 mutated (140), and CEBPA mutated (141). Currently, enrollment in the phase 1 dose escalation study is on-going; a phase 2 expansion study in r/r NPM1 mutated and KMT2Ar AML is planned following the determination of the recommended phase 2 dose.

A second structurally unrelated menin inhibitor, VTP50469, was discovered via iterative structure based design (142). Similar to the MI series of compounds, VTP50469 binds to the KMT2A binding pocket of menin but with a distinct binding mode that engages somewhat different amino acids. VTP50469 inhibits the growth of KMT2Ar leukemia cells, reduces KMT2A fusion protein target gene expression, improved survival in a murine MLL-AF9 leukemia model and in KMT2Ar PDX, with some PDX mice surviving over 1 year (142). Subsequently, VTP50469 was also shown to reduce HOX gene expression and inhibit in vivo growth in NPM1 mutated murine cells as well as NPM1 mutated AML PDX mice (125). An analog of VTP50469, SYNDX-5613, is currently being investigated in a phase 1/2 study in r/r AML (NCT04065399). The phase 1 dose escalation study does not require any specific mutation or chromosome re-arrangement, while the planned phase 2 expansion will include KMT2Ar AML, KMT2Ar acute lymphoblastic leukemia (ALL) and NPM1 mutated AML. Preliminary results were reported at the 2020 AACR meeting (143). Of 6 evaluable patients, two had objective responses, including a CRi in a t(10;11), FLT3-ITD AMl patient and a partial response after 28 days of therapy in a t(9;11) AML patient. Of note, this patient continues on study. No DLTs have been identified but two patients had grade 1–2 QTc prolongation that resolved with dose reductions. In addition, unlike KO-539, the pharmacokinetics of SYNDX-5613 appeared affected by CYP3A4 inhibitors, with patients taking concurrent CYP3A4 inhibitors more likely to reach target plasma concentrations greater than the predicted IC95 for the menin-KMT2A interaction. Based on these preliminary results, SYNDX-5613 was granted orphan drug designation by the FDA in April 2020.

Conclusions and Future Directions

Since 2017, eight new drugs have been approved for the treatment of AML. Our understanding of the altered signaling pathways and epigenetic perturbations underlying leukemia cell survival and proliferation continues to grow. With this new knowledge, additional potential pharmacologic targets are identified and novel targeted inhibitors developed, including drugs designed to inhibit pathways previously considering un-druggable. However, our excitement is tempered by the knowledge that AML remains an incurable disease for the majority of patients, especially older patients and those with medical co-morbidities limiting treatment. We now have an “embarrassment of riches” when it comes to potential novel therapeutics for AML. As of February 2021, 497 actively recruiting clinical trials for adult AML are currently registered with clinicaltrials.gov. The challenge for the field will be to identify the most promising new drugs, impact for particular genomic subgroups and determine the most optimal combinations or sequences of novel therapeutics.

Acknowledgments

Funding: No funding was received.

Footnotes

Disclosures

Compliance with Ethical Standards

Conflict of Interest

Dr. Nicole Grieselhuber declares that she has no conflict of interest.

Dr. Alice Mims has served on the advisory boards for Syndax Pharmaceuticals, Kura Oncology, Jazz Pharmaceuticals, and AbbVie Pharmaceuticals.

Human and Animal Rights and Informed Consent

This article contains no studies with human or animal subjects performed by either of the authors.

Publisher's Disclaimer: This Author Accepted Manuscript is a PDF file of a an unedited peer-reviewed manuscript that has been accepted for publication but has not been copyedited or corrected. The official version of record that is published in the journal is kept up to date and so may therefore differ from this version.

References

Papers of particular interest have been highlighted as:

• Of importance

•• Of major importance

  • 1.Döhner H, Weisdorf DJ, Bloomfield CD. Acute Myeloid Leukemia. N Engl J Med 2015:373:1136–52. Doi: 10.1056/NEJMra1406148. [DOI] [PubMed] [Google Scholar]
  • 2.Ley TJ, Miller C, Ding L, Raphael BJ, Mungall AJ, Robertson AG, et al. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med 2013:368:2059–74. Doi: 10.1056/NEJMoa1301689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Papaemmanuil E, Gerstung M, Bullinger L, Gaidzik VI, Paschka P, Roberts ND, et al. Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med 2016:374:2209–21. Doi: 10.1056/NEJMoa1516192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Eisfeld AK, Mrόzek K, Kohlschmidt J, Nicolet D, Orwick S, Walker CJ, et al. The mutational oncoprint of recurrent cytogenetic abnormalities in adult patients with de novo acute myeloid leukemia. Leukemia 2017:31:2211–18. Doi: 10.1038/leu.2017.86 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Stein EM, DiNardo CD, Pollyea DA, Fathi AT, Roboz GJ, Altman JK, et al. Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia. Blood 2017:130:722–31. Doi: 10.1182/blood-2017-03-779405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Pollyea DA, Tallman MS, de Botton S, Kantarjian HM, Collins R, Stein AS, et al. Enasidenib, an inhibitor of mutant IDH2 proteins, induces durable remissions in older patients with newly diagnosed acute myeloid leukemia. Leukemia 2019:33:2575–84. Doi: 10.1038/s41275-019-0472-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.DiNardo CD, Stein EM, doBotton S, Roboz GJ, Altman JK, Mims AS, et al. Durable remissions with ivosidenib in IDH1-mutated relapsed or refractory AML. N Engl J Med 2018:378:2386–98. Doi: 10.1056/NEJMoa1716984. [DOI] [PubMed] [Google Scholar]
  • 8.Roboz GJ, DiNardo CD, Stein EM, de Botton S, Mims AS, Prince GT, et al.Ivosidenib induces deep durable remissions in patients with newly diagnosed IDH1-mutant acute myeloid leukemia. Blood 2020:135:463–71. Doi: 10.1182/blood.2019002140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Stone RM, Mandrekar SJ, Sanford BL, Laumann K, Geyer S, Bloomfield CD, et al. Midostaurin plus chemotherapy for acute myeloid leukemia with a FLT3 mutation. N Engl J Med 2017:377:454–64. Doi: 10.1056/NEJMoa1614359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Perl AE, Martinelli G, Cortes JE, Neubauer A, Berman E, Paolini S, et al. Gilteritinib or chemotherapy for relapsed or refractory FLT3-mutated AML. N Engl J Med 2019:381:1728–40. Doi: 10.1056/NEJMoa1902688. [DOI] [PubMed] [Google Scholar]
  • 11.Wei AH, Montensinos P, Ivanov V, DiNardo CD, Novak J, Laribi K, et al. Venetoclax plus LDAC for newly diagnosed AML ineligible for intensive chemotherapy: a phase 3 randomized placebo-controlled trial. Blood 2020:135:2137–45. Doi: 10.1182/blood.2020004856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.DiNardo CD, Jonas BA, Pullarkat V, Thirman MJ, Garcia JS, et al. Azacitidine and venetoclax in previously untreated 2020:383:617–29. Doi: 10.1056/NEJMoa2012971. [DOI] [PubMed] [Google Scholar]
  • 13.Cortes JE, Heidel FH, Hellmann A, Fiedler W, Smith BD, Robak T, et al. Randomized comparison of low dose cytarabine with or without glasdegib in patients with newly diagnosed acute myeloid leukemia or high-risk myelodysplastic syndrome. Leukemia 2019:33:379–89. Doi: 10.1038/s41375-018-0312-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Zuber J, Shi J, Wang E, Rappaport AR, Herrmann H, Sison EA, et al. RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukemia. Nature 2011:478:524–8. Doi: 10.1038/nature10334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Herrmann H, Blatt K, Shi J, Gleixner KV, Cerny-Reiterer S, Mȕllauer L, et al. Small-molecule inhibition of BRD4 as a new potent approach to eliminate leukemic stem- and progenitor cells in acute myeloid leukemia AML. Oncotarget 2012:3(12):1588–99. Doi: 10.18632/oncotarget.733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Pericole FV, Lazarini M, de Paiva LB, Duarte ADSS, Ferro KPV, Niemann FS, et al. BRD4 inhibition enhances azacitidine efficacy in acute myeloid leukemia and myelodysplastic syndromes. Front Oncol 2019:9:16. Doi: 10.3389/fonc.2019.00016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Tzelepis K, De Braekeleer E, Aspris D, Barbieri I, Vijayabaskar MS, Liu WH, et al. SRPK1 maintains acute myeloid leukemia through effects on isoform usage of epigenetic regulators including BRD4. Nat Comm 2018:9:5378. Doi: 10.1038/s41467-018-07620-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Roe JS, Vakoc CR. The essential transcriptional function of BRD4 in acute myeloid leukemia. Cold Spring HarbSymp Quant Biol 2016:81:61–66. Doi: 10.1101/sqb.2016.81.031039. •An excellent review of the function of BRD4 in AML
  • 19.Dawson MA, Prinjha RK, Dittmann A, Giotopoulos G, Bantscheff M, Chan WI, et al. Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia. Nature 2011:478:52933. Doi: 10.1038/nature10509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Delmore JE, Issa GC, Lemieux ME, Rahl PB, Shi J, Jacob HM, et al. BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 2011:146:904–17. Doi: 10.1016/j.cell.2011.08.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Dawson MA, Gudgin EJ, Horton SJ, Giotopoulos G, Meduri E, Robson S, et al. Recurrent mutations, including NPM1c, activate a BRD4-dependent core transcriptional program in acute myeloid leukemia. Leukemia 2014:28:311–20. Doi: 10.1038/leu.2013.338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Chen C, Liu Y, Lu C, Cross JR, Morris JP 4th, Shroff AS, et al. Cancer-associated IDH2 mutants drive an acute myeloid leukemia that is susceptible to Brd4 inhibition. Genes Dev 2013:27:2974–85. Doi: 10.1101/gad.226613.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Zhang P, He F, Bai J, Yamamoto S, Chen S, Zhang L, et al. Chromatin regulator Asxl1 loss and Nf1 haploinsufficiency cooperate to accelerate myeloid malignancy. J Clin Invest 2018:128:5383–98. Doi: 10.1172/JCI121366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Zhao Y, Liu Q, Acharya P, Stengel KR, Sheng Q, Zhou X, et al. High-resolution mapping of RNA polymerases identifies mechanisms of sensitivity and resistance to BET inhibitors in t(8;21) AML. Cell Rep 2016:16:2003–16. Doi: 10.1016/j.celrep.2016.07.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Saenz DT, Fiskus W, Manshouri T, Rajapakshe K, Krieger S, Sun B, et al. BET protein bromodomain inhibitor-based combinations are highly active against post-myeloproliferative neoplasm secondary AML cells. Leukemia 2017:31:678–87. Doi: 10.1038/leu.2016.260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Petretich M, Demont E, Grandi P, et al. Domain-selective targeting of BET proteins in cancer and immunological diseases. 2020:S1367–5931:30019–3. Doi: 10.1016/j.cbpa.2020.02.003. [DOI] [PubMed] [Google Scholar]
  • 27.Tyler DS, Vappiani J, Cañeque T, Lam EYN, Ward A, Gilan O, et al. Click chemistry enables preclinical evaluation 2017:356:1397–1401. Doi: 10.1126/science.aal2066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Gilan O, Rioja I, Knezevic K, Bell MJ, Yeung MM, Harker NR, et al. Selective targeting of BD1 and BD2 of the BET proteins in cancer and immunoinflammation. Science 2020:368:387–94. Doi: 10.1126/science.aaz8455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Berthon C, Raffoux E, Thomas X, Vey N, Gomez-Roca C, Yee K, et al. Bromodomain inhibitor OTX015 in patients with acute leukaemia: a dose-escalation, phase 1 study. Lancet Haematol 2016: 3:e186–95. Doi: 10.1016/S2352-3026(15)00247-1. ••First clinical trial of a BET inhibitor in acute leukemia
  • 30. Amorim S, Stathis A, Gleeson M, Iyengar S, Magarotto V, Leleu X, et al. Bromodomain inhibitor OTX015 in patients with lymphoma or multiple myeloma: a dose-escalation, open-label, pharmacokinetic, phase 1 study. Lancet Haematol 2016:3:E196–204. Doi: 10.1016/S2452-3026(16)00021-1. •The phase 1 study of the BET inhibitor OTX015 in lymphoid malignancies, a companion study to reference 29 above
  • 31.Yang L, Ding L, Liang J, Chen J, Tang Y, Xue H, et al. Relatively favorable prognosis for MLL-rearranged childhood acute leukemia with reciprocal translocations. Ped Blood Cancer 2018:65:e27266. Doi: 10.1002/pbc.27266. [DOI] [PubMed] [Google Scholar]
  • 32.Super HJ, McCabe NR, Thirman MJ, Larson RA, Le Beau MM, Pedersen-Bjergaard J, et al. Rearrangements of the MLL gene in therapy-related acute myeloid leukemia in patients previously treated with agents targeting DNA-topoisomerase II. Blood 1993:83:3705–11. [PubMed] [Google Scholar]
  • 33.Chen Y, Kantarjian H, Pierce S, Faderl S, O’Brien S, Qiao W, et al. Prognostic significance of 11q23 aberrations in adult acute myeloid leukemia and the role of allogeneic stem cell transplantation. Leukemia 2013:27:836–42. Doi: 10.1038/leu.2012.319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Uckelmann HJ, Armstrong SA. Chromatin complexes maintain self-renewal of myeloid progenitors in AML: opportunities for therapeutic intervention. Stem Cell Reports 2020:15:6–12. Doi: 10.1016/j.stemcr.2020.05.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Bernt KM, Zhu N, Sinha AU, Vempati S, Faber J, Krivtsov AV, et al. MLL-rearranged leukemia is dependent on aberrant H3K79 methylation by DOT1 L. Cancer Cell 2011:20L66–78. Doi: 10.1016/j.ccr.2011.06.010. •Demonstration of the dependence of MLLr AML on DOT1L
  • 36. Daigle SR, Olhava EJ, Therkelsen CA, Majer CR, Sneeringer CJ, Song J, et al. Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor. Cancer Cell 2011:20:53–65. Doi: 10.1016/j.ccr.2011.06.009. •Pre-clinical development of a DOT1L inhibitor
  • 37.Meyer C, Burmeister T, Gröger D, Tsaur G, Fechina L, Renneville A, et al. The MLL recombinome of acute leukemias in 2017. Leukemia 2018:32:273–84. Doi: 10.1038/leu.2017.213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Daigle SR, Olhava EJ, Therkelsen CA, Basavapathruni A, Jin L, Boriack-Sjodin PA, et al. Potent inhibition of DOT1L as treatment of MLL-fusion leukemia. Blood 2013:122:1017–25. Doi: 10.1182/blood-2013-04-497644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Stein EM, Garcia-Manero G, Rizzieri DA, Tibes R, Berdeja JG, Savona MR, et al. The DOT1L inhibitor pinometostat reduces H3K79 methylation and has modest clinical activity in adult acute leukemia. Blood 2018:131:2661–9. Doi: 10.1182/blood-2017-09-806679, ••Initial phase 1 study of pinometostat in acute leukemia
  • 40.Shukla N, Wetmore C, O’Brien MM, Silverman LB, Brown P, Cooper TM, et al. Final report of phase 1 study of the DOT1L inhibitor, pinometostat (EPZ-5676), in children with relapsed or refractory MLL-r acute leukemia. Blood 2016:128: Abstract Doi: 10.1182/blood.V128.22.2780.2780. [DOI] [Google Scholar]
  • 41. Goroschuk O, Kolosenko I, Vidarsdottir L, Azimi A, Palm-Apergi C. Polo-like kinases and acute leukemia. Oncogene 2019:38:1–16. Doi: 10.1038/s41388-018-0443-5. •An excellent review on the role of polo-like kinases in leukemia
  • 42.Liu X. Targeting polo-like kinases: a promising therapeutic approach for cancer treatment. Transl Oncol 2015:8:185–95. Doi: 10.1016/j.tranon.2015.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Renner AG, Santos CD, Recher C, Bailly C, Créancier L, Kruczynski A, et al. Polo-like kinase 1 is overexpressed in acute myeloid leukemiaand its inhibition preferentially targets the proliferation of leukemic cells. Blood 2009:114:659–62. Doi: 10.1182/blood-208-12-195867. [DOI] [PubMed] [Google Scholar]
  • 44.Tao YF, Li ZH, Du WW, Xu LX, Ren JL, Li XL, et al. Inhibiting PLK1 induces autophagy of acute myeloid leukemia cells via mammalian target of rapamycin pathway dephosphorylation. Oncol Rep 2017:37:1419–29. Doi: 10.3892/or.2017.5417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Ikezoe T, Yang J, Nishioka C, Takezaki Y, Tasaka T, Togitani K, et al. A novel treatment strategy targeting polo-like kinase 1 in hematological malignancies. Leukemia 2009:29:1564–76. Doi: 10.1038/leu.2009.94. [DOI] [PubMed] [Google Scholar]
  • 46.Steegmaier M, Hoffmann M, Baum A, Lénárt P, Petronczki M, Krssák M, et al. BI 2536, a potent and selective inhibitor of polo-like kinase 1, inhibits tumor growth in vivo. CurrBiol 2007:17:316–22. Doi: 10.1016/j.cub.2006.12.037. [DOI] [PubMed] [Google Scholar]
  • 47. Moison C, Lavallée P, Thiollier C, Lehnertz B, Boivin I, Mayotte N, et al. Complex karyotype AML displays G2/M signature and hypersensitivity to PLK1 inhibition. Blood Advances 2019:3:552–63. Doi: 10.1182/bloodadvances.2018.028480. •Evidence for a PLK1 dependence in complex karyotype AML
  • 48.Simonetti G, Padella A, do Valle IF, Fontana MC, Fonzi E, Bruno S, et al. Aneuploid acute myeloid leukemia exhibits a signature of genomic alterations in the cell cycle and protein degradation machinery. Cancer 2019:126:712–25. Doi: 10.1002/cncr.31837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Dill V, Kauschinger J, Hauch RT, Buschhorn L, Odinius TO, Mȕller-Thomas C, et al. Inhibition of PLK1 by capped-dose volasertib exerts substantial efficacy in MDS and sAML while sparing healthy haematopoiesis. Eur J Haematol 2020. ”104:125–37. Doi: 10.1111/ejh.13354. [DOI] [PubMed] [Google Scholar]
  • 50. Müller-Tidow C, Bug G, Lübbert M, Krämer A, Krauter J, Valent P, et al. A randomized, open-label, phase I/II trial to investigate the maximum tolerated dose of the polo-like kinase inhibitor BI 2536 in elderly patients with refractory/relapsed acute myeloid leukaemia. Br J Haematol 2013:163:214–22. Doi: 10.1111/bjh.12518. ••The first study of a PLK inhibitor in AML
  • 51.Gjertsen BT, Schöffski P. Discovery and development of the polo-like kinase inhibitor volasertib in cancer therapy. Leukemia 2015:29:11–9. Doi: 10.1038/leu.2014.222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Rudolph D, Steegmaier M, Hoffmann M, Grauert M, Baum A, Quant J, et al. BI 6727, a polo-like kinase inhibitor with improved pharmacokinetic profile and broad antitumor activity. Clin Cancer Res 2009:15:3094–102. Doi: 10.1158/1078-0432.CCR-08-2445. [DOI] [PubMed] [Google Scholar]
  • 53.Ciceri P, Müller S, O’Mahony A, Fedorov O, Filippakopoulos P, Hunt JP, et al. Dual kinase-bromodomain inhibitors for rationally designed polypharmacology. Nat Chem Biol 2014:10:305–12. Doi: 10.1038/nchembio.1471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Schöffski P, Awada A, Dumez H, Gil T, Bartholomeus S, Wolter P, et al. A phase I, dose-escalation study of the novel polo-like kinase inhibitor volasertib (BI 9727) in patients with advanced solid tumours. Eur J Cancer 2012:48:179–86. Doi: 10.1016/j.ejca.2011.11.001. [DOI] [PubMed] [Google Scholar]
  • 55.Stadler WM, Vaughn DJ, Sonpavde G, Vogelzang NJ, Tagawa ST, Petrylak DP, et al. An open-label, single-arm, phase 2 trial of the polo-like kinase inhibitor volasertib (BI 6727) in patients with locally advanced or metastatic urothelial cancer. Cancer 2014:120:976–82. Doi: 10.1002/cncr.28519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Lin CC, Su WC, Yen CJ, Hsu CH, Su WP, Yeh KH, et al. A phase I study of two dosing schedules ofvolasertib (BI 6727), an intravenous polo-like kinase inhibitor, in patients with advanced solid malignancies. Br J Cancer 2014:110:2434–40. Doi: 10.1038/bjc.2014.195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Ottmann OG, Müller-Tidow C, Krämer A, Schlenk RF, Lübbert M, Bug G, et al. Phase I dose-esclation trial investigating volasertib as monotherapy or in combination with cytarabine in patients with relapsed/refractory acute myeloid leukaemia. Br J Haematol 2019:184:1018–21. Doi: 10.1111/bjh.15204. ••Phase 1 study of volasertib in r/r AML showing safety as monotherapy and when combined with LDAC
  • 58.Döhner H, Lübbert M, Fiedler W, Fouillard L, Haaland A, Brandwein JM, et al. Randomized, phase 2 trial of low-dose cytarabine with or without volasertib in AML patients not suitable for induction therapy. Blood 2014:124:1426–33. Doi: 10.1182/blood-2014-03-560557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Döhner H, Symeonidis A, Sanz AM, Deeren D, Demeter J, Anagnostopoulos A, et al. Phase III randomized trial of volasertib plus low-dose cytarabine (LDAC) versus placebo plus LDAC in patients aged ≥65 years with previously untreated AML, ineligible for intensive therapy. European Hematology Association 2016 Annual Meeting. Abstract S501. [Google Scholar]
  • 60.Cortes J, Podoltsev N, Kantarjian H, Borthakur G, Zeidan AM, Stahl M, et al. Phase 1 dose escalation trial of volasertib in combination with decitabine in patients with acute myeloid leukemia. Int J Hematol 2021:113:92–99. Doi: 10.1007/s12185-020-02994-8. [DOI] [PubMed] [Google Scholar]
  • 61.Benetatos L, Dasoula A, Hatzimichael E, Syed N, Voukelatou M, Dranitsaris G, et al. Polo-like kinase 2 (SNK/PLK2) is a novel epigenetically regulated gene in acute myeloid leukemia and myelodysplastic syndromes: genetic and epigenetic interactions. Ann Hematol 2011:90:1037–45. Doi:. [DOI] [PubMed] [Google Scholar]
  • 62.Deng S, Wang H, Jia C, Zhu S, Chu X, Ma Q, et al. MicroRNA-146a induces linage-negative bone marrow cell apoptosis and senescence by targeting polo-like kinase 2 expression. Arterioscler Thromb Vasc Biol 2017:37:280–90. Doi: 10.1161/ATVBAHA.116.308378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Beria I, Bossi RT, Brasca MG, Caruso M, Ceccarelli W, Fachin G, et al. NMS-P937, a 4,5-dihydro-1H-pyrazolo[4,3-h] quinazoline derivative as potent and selective polo-like kinase 1 inhibitor. Bioorg Med Chem Lett 2011:21:2969–74. Doi: 10.1016/j.bmcl.2011.03.054. [DOI] [PubMed] [Google Scholar]
  • 64.Valsasina B, Beria I, Alli C, Alzani R, Avanzi N, Ballinari D, et al. NMS-P937, an orally available, specific small-molecule polo-like kinase 1 inhibitor with antitumor activity in solid and hematologic malignancies. Mol Cancer Ther 2012:11:1006–16. Doi: 10.1158/1535-7613.MCT-11-0765. [DOI] [PubMed] [Google Scholar]
  • 65. Weiss GJ, Jameson G, Von Hoff DD, Valsasina B, Davite C, Giulio CD, et al. Phase I dose escalation study of NMS-1286937, an orally available polo-like kinase 1 inhibitor, in patients with advanced or metastatic solid tumors. Invest New Drugs 2018:36:85–95. Doi: 10.1007/s10637-017-0491-7. ••First clinical trial of onvansertib, which has improved PLK1 specificity
  • 66. Zeidan AM, Ridinger M, Lin TL, Becker PS, Schiller GJ, Patel PA, et al. A phase Ib study of onvansertib, a novel oral PLK1 inhibitor, in combination therapy for patients with relapsed or refractory acute myeloid leukemia. Clin Cancer Res 2020:26:6132–6140. doi: 10.1158/1078-0432.CCR-20-2586. ••Clinical trial of onvansertib in AML
  • 67.Zeidan AM, Lin T, Becker PS, Schiller GJ, Patel PA, Spira AI, et al. Updated analysis of a phase 1b/2 study of onvansertib, a PLK1 inhibitor, in combination with decitabine in patients with relapsed or refactory acute myeloid leukemia. American Society of Hematology 2020 Annual Meeting: Abstract 1954. [Google Scholar]
  • 68.Athuluri-Divakar SK, Vasquez-Del Carpio R, Dutta K, Baker SJ, Consenza SC, et al. A small molecule RAS-mimetic disrupts RAS association with effector proteins to block signaling. Cell 2016:165:643–55. Doi: 10.1016/j.cell.2016.03.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Duffy MJ, Synnott NC, O’Grady S, Crown J. Targeting p53 for the treatment of cancer. Seminars Cancer Biol 2020:30LS1044–579X(20)30160–7. Doi: 10.1016/j.semcancer.2020.07.005. [DOI] [PubMed] [Google Scholar]
  • 70.Wong TN, Ramsingh R, Young AL, Miller CA, Touma W, Welch JS, et al. Role of TP53 mutations in the origin and evolution of therapy-related acute myeloid leukaemia. Nature 2014:518:552–5. Doi: 10.1038/nature13968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Grinfeld J, Nangalia J, Baxter EJ, Wedge DC, Angelopoulos N, Cantrill R, et al. Classification and personalized prognosis in myeloproliferative neoplasms. N Engl J Med 2018:379:1416–30. Doi: 10.1056/NEJMoa1716614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Mrózek K, Eisfeld AK, Kohlschmidt J, Carroll AJ, Walker CJ, Nicolet D, et al. Complex karyo type inde novo acute myeloid leukemia: typical and atypical subtypes differ molecularly and clinically. Leukemia 2019:33:1620–34. Doi: 10.1038/s41375-019-0390-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Rucker FG, Schlenk RF, Bullinger L, Kayser S, Teleanu V, Kett H, et al. TP53 alterations in acute myeloid leukemia with complex karyotype correlate with specific copy number alterations, monosomal karyotype, and dismal outcome. Blood 2012:119:2114–21. Doi: 10.1182/blood-2011-08-3715758. [DOI] [PubMed] [Google Scholar]
  • 74.Boettcher S, Miller PG, Sharma R, McConkey M, Leventhal M, Krivstov A, et al. A dominant-negative effect drives selection of TP53 missense mutations in myeloid malignancies. Science 2019:365:599–604. Doi: 10.1126/science.aax3649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Mims AS, Kohlschmidt J, Eisfeld AK, Mrózek K, Blachly JS, Orwick S, et al. Comparison of clinical and molecular characteristics of patients with acute myeloid leukemia and either TP73 or TP53 mutations. Leukemia 2020:Aug 5.Epub.Online ahead of print. DOi: 10.1038/s41375-020-1007-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Prochazka KT, Pregartner G, Rücker FG, Heitzer E, Pabst G, Wölfer A, et al. Clinical implications ofsubclonal TP53 mutations in acute myeloid leukemia. Haematologica 2019:104:516–23. Doi: 10.334/haematol.2018.205013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Welch JS, Pett AA, Miller CA, Fronick CC, O’Laughlin M, Fulton RS, et al. TP53 and decitabine in acute myeloid leukemia and myelodysplastic syndromes. N Engl J Med 2016:375:2023–36. Doi: 10.1056/NEJMoa1605949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Becker H, Pfeifer D, Ihorst G, Pantic M, Wehrle J, Rüter BH, et al. Monosomal karyotype and chromosome 17p loss or TP53 mutations in decitabine-treated patients with acute myeloid leukemia. Ann Hematol 2020:99:1551–60. Doi: 10.1007/s00277-020-04082-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Short NJ, Kantarjian HM, Loghavi S, Huang X, Qiao W, Borhakur G, et al. Treatment with a 5-day versus a 10-day schedule of decitabine in older patients with newly diagnosed acute myeloid leukaemia: a randomized phase 2 trial. Lancet Haematol 2019:6:e29–e37. Doi: 10.1016/S2352-2036(1)30182-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Lambert JMR, Gorzov P, Veprintsev DB, Söderqvist, Segerbäck D, Bergman J, et al. PRIMA-1 reactivates mutant p53 by covalent binding to the core domain. Cancer Cell 2009:15:376–88. Doi: 10.1016/j.ccr.2009.03.003. [DOI] [PubMed] [Google Scholar]
  • 81. Zhang Q, Bykov VJN, Wiman KG, Zawacka-Pankau J, et al. APR-246 reactivates mutant p53 by targeting cysteines 124 and 277. Cell Death Dis 2018:9:439. Doi: 10.1038/s41419-018-0463-7. •Description of the mechanism by which APR-246 converts mutated p53 to wild-type activity
  • 82.Tessoulin B, Descamps G, Moreau P, Maïga S, Lodé L, Godon C, et al. PRIMA-1met induces myeloma cell death independent of p53 by impairing the GSH/ROS balance. Blood 2014:124:1626–36. Doi: 10.1182/blood-2014-01-548800. [DOI] [PubMed] [Google Scholar]
  • 83.Maslah N, Salomao N, Drevon L, Verger E, Partouche N, Ly P, et al. Synergistic effects of PRIMA-1Met (APR-246) and 5-azacitidine in TP53-mutated myelodysplastic syndromes and acute myeloid leukemia. Haematologica 2020:105L1539–51. Doi: 10.3324/haematol.2019.218453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Lehman S, Bykov VJN, Ali D, Andrén O, Cherif H, Tidefelt U, et al. Targeting p53 in vivo: a first-in-human study with p53-targeting compound APR-246 in refractory hematologic malignancies and prostate cancer. J Clin Oncol 2012:30:3633–9. Doi: 10.1200/JCO.2011.40.7783. ••First-in-man study of a mutant TP53 inhibitor
  • 85. Deneberg S, Cherif H, Lazarevic V, Andersson P-O, von Euler M, Juliusson G, et al. An open-label phase I dose-finding study of APR-246 in hematological malignancies. Blood Cancer J 2016:6:e447. Doi: 10.1038/bcj.2016.60. ••Optimization of infusion protocol allowed for increased APR-246 MTD in hematologic malignancies
  • 86. Sallman DA, DeZern AE, Garcia-Manero G, Steensma DP, Roboz GJ, Sekeres MA, et al. Eprenetapopt (APR-246) and azacitidine in TP53-mutant myelodysplastic syndromes. J Clin Oncol 2021. Doi: 10.1200/JCO.20.02341. Online ahead of print. ••Phase 1 study of APR-146 in TP53 mutated myeloid malignancies
  • 87. Cluzeau T, Sebert M, Rahmé R, Cuzzubo S, Lehmann-Che J, Madelaine I, et al. Eprenetapopt plus azacitidine in TP53-mutated myelodysplastic syndromes and acute myeloid leukemia: a phase II study by the Groupe Francophone des Myélodysplasies (GFM). J Clin Oncol 2021. Doi: 10.1200/JCO.20.02342. Online ahead of print. ••Phase 2 study of APR-146 in TP53 mutated myeloid malignancies
  • 88.Vassilev LT, Vu BT, Graves B, Carvajal D, Podlaski F, Filipovic Z, et al. In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science 2004:303:844–8. [DOI] [PubMed] [Google Scholar]
  • 89. Andreeff M, Kelly KR, Yee K, Assouline S, Strair R, Popplewell L, et al. Results of the phase 1 trial of RG7112, a small-molecule MDM2 antagonist in leukemia. Clin Cancer Res 2016:22:868–76. Doi: 10.1158/1078-0432.CCR-15-0481. ••First phase 1 study of a MDM2 inhibitor in leukemia
  • 90.Ray-Coquard I, Blay JY, Italiano A, Cesne AL, Penel N, Zhi J, et al. Effect of the MDM2 antagonist RG7112 on the p53 pathway in patients with MDM2-amplified, well-differentiated or dedifferentiated liposarcoma: an exploratory proof-of-mechanism study. Lancet Oncol 2012:13:1133–40. Doi: 10.1016/S1470-2045(12)70474-6. [DOI] [PubMed] [Google Scholar]
  • 91.Ding Q, Zhang Z, Liu JJ, Jiang N, Zhang J, Ross TM, et al. Discovery of RG7388, a potent and selective p53-MDM2 inhibitor in clinical development. J Med Chem 2013:56:5979–83. Doi: 10.1021/jm400487c. [DOI] [PubMed] [Google Scholar]
  • 92.Yee K, Papayannidis C, Vey N, Dickinson MJ, Kelly KR, Assouline S, et al. Murine double minute 2 inhibition alone or with cytarabine in acute myeloid leukemia: Results from an idasanutlin phase 1/1b study.Leuk Res 2021. Doi: 10.1016/j.leukres.2020.106489. Epub 2020 Dec 1. [DOI] [PubMed] [Google Scholar]
  • 93.Reis B, Jukofsky L, Chen G, Martinelli G, Zhong HSo WV, et al. Acute myeloid leukemia patients’ clinical response to idasanutlin (RG7388) is associated with pre-treatment MDM2 protein expression in leukemic blasts. Haematologica 2016:101:e185–8. Doi: 10.3324/haematol.2015.139717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Mascarenhas J, Lu M, Kosiorek H, Virtgaym E, Xia L, Sandy L, et al. Oral idasanutlin in patients with polycythemia vera. Blood 2019:134:525–33. Doi: 10.1182/blood.2018893545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Montesinos P, Beckermann BM, Catalani O, Esteve J, Gamel K, Konopleva MY, et al. MIRROS: a randomized, placebo-controlled, Phase III trial of cytarabine ± idasanutlin in relapsed or refractory acute myeloid leukemia. Future Oncol 2020:16:807–815. Doi: 10.2217/fon-2020-0044. [DOI] [PubMed] [Google Scholar]
  • 96.Siu LL, Italiano A, Miller WH, Blay JY, Gietema JA, Bang YJ, et al. Phase 1 dose escalation, food effect, and biomarker study of RG7388, a more potent second-generation MDM2 antagonist, in patients (pts) with solid tumors. J Clin Oncol 2014:32:2535. Doi: 10.1200/jco.2014.32.15_suppl.2535. [DOI] [Google Scholar]
  • 97.Nemunaitis J, Young A, Ejadi S, Miller W, Chen LC, Nichols G, et al. Effects of posaconazole (a strong CYP3A4 inhibitor), two new tablet formulations, and food on the pharmacokinetics of idasanutlin, an MDM2 antagonist, in patients with advanced solid tumors. Cancer Chemother Pharmacol 2018:81:529–37. Doi: 10.1007/s00280-018-3521-z. [DOI] [PubMed] [Google Scholar]
  • 98.Higgins B, Glenn K, Walz A, Tovar C, Filipovic Z, Hussain S, et al. Preclinical optimization of MDM2 antagonist scheduling for cancer treatment by using a model-based approach. Clin Cancer Res 2014:20:3742–52. Doi: 10.1158/1078-0432.CCR-14-0460. [DOI] [PubMed] [Google Scholar]
  • 99.Daver NG, Garcia JS, Jonas BA, Kelly KR, Assouline S, Brandwein JM, et al. Updated results from the venetoclax (Ven) in combination with idasanutlin (Idasa) arm of a phase 1b trial in elderly patients with relapsed or refractory (R/R) AML ineligible for cytotoxic chemotherapy. Blood 2019:134:229. [Google Scholar]
  • 100.Rew Y and Sun D. Discovery of a small molecule MDM2 inhibitor (AMG 232) for treating cancer. J Med Chem 2014:57:6332–41. Doi: 10.1021/jm500627s. [DOI] [PubMed] [Google Scholar]
  • 101.Gluck WL, Gounder MM, Frank R, Eskens F, Blay JY, Cassier PA, et al. Phase 1 study of the MDM2 inhibitor AMG 232 in patients with advanced p53 wild-type solid tumors or multiple myeloma. Invest New Drugs 2020:38:831–43. Doi: 10.1007/s10637-019-00840-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Erba HP, Becker PS, Shami PJ, Grunwald MR, Flesher DL, Zhu M, et al. Phase 1b study of the MDM2 inhibitor AMG 232 with or without trametinib in relapsed/refractory acute myeloid leukemia. Blood Adv 2019:3:1393–1949. Doi: 10.1182/bloodadvances.2019030916. ••Phase 1 study showing safety and evidence for activity of AMG-232 in AML
  • 103.Carvajal LA, Neriah DB, Senecal A, Benard L, Thiruthuvanathan V, Yatsenko T, et al. Dual inhibition of MDMX and MDM2 as a therapeutic strategy in leukemia. Science Transl Med 2018:10:eaao3003. Doi: 10.1126/scitranslmed.aao3003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Sallman DA, Borate U, Cull EH, Donnellan WB, Komrokji RS, Steidl UG, et al. Phase 1/1b study of the stapled peptide ALRN-6924, a dual inhibitor of MDMX and MDM2, as monotherapy or in combination with cytarabine for the treatment of relapsed/refractory AML and advanced MDS with TP53 wild-type. Blood 2018:132:4066. Doi: 10.1182/blood-2018-99-118780. [DOI] [Google Scholar]
  • 105.Armstrong SA, Staunton JE, Silverman LB, Pieters R, den Boer ML, Minden MD, et al. MLL translocations specify a distinct gene expression profile that distinguishes a unique leukemia. Nat Genet:30:41–47. Doi: 10.1038/ng765. [DOI] [PubMed] [Google Scholar]
  • 106.Hinai ASAA, Pratcorona M, Grob T, Kavelaars FG, Bussaglia E, Sanders MA, et al. The landscape of KMT2A-PTD AML: Concurrent mutations, gene expression signatures, and clinical outcome. Hemasphere 2019:3:e181. Doi: 10.1097/HS9.0000000000000181 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Alcalay M, Tiacci E, Bergomas R, Bigerna B, Venturini E, Minardi SP, et al. Acute myeloid leukemia bearing cytoplasmic nucleophosmin (NPMc+ AML) shows a distinct gene expression profile characterized by up-regulation of genes involved in stem-cell maintenance. Blood 2005:106:899–902. Doi: 10.1182/blood-2005-02-0560. [DOI] [PubMed] [Google Scholar]
  • 108.Mullighan CG, Kennedy A, Zhou X, Radtke I, Phillips LA, Shurtleff SA, et al. Pediatric acute myeloid leukemia with NPM1 mutations is characterized by a gene expression profile with dysregulated HOX gene expression distinct from MLL-rearranged leukemias. Leukemia. 2007:9:2000–2009. doi: 10.1038/sj.leu.2404808. [DOI] [PubMed] [Google Scholar]
  • 109.Spencer DH, Young MA, Lamprecht TL, Helton NM, Fulton R, O’Laughlin M, et al. Epigenomic analysis of the HOX gene loci reveals mechanisms that may control canonical expression patterns in AML and normal hematopoietic cells. Leukemia. 2015:6:1279–1289. doi: 10.1038/leu.2015.6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Caligiuri MA, Strout MP, Lawrence D, Arthur DC, Baer MR, Yu F, Knuutila S, et al. Rearrangement of ALL1 (MLL) in acute myeloid leukemia with normal cytogenetics. Cancer Res. 1998:58:55–59. [PubMed] [Google Scholar]
  • 111.Döhner K, Tobis K, Ulrich R, Fröhling S, Benner A, Schlenk RF, et al. Prognostic significance of partial tandem duplications of the MLL gene in adult patients 16 to 60 years old with acute myeloid leukemia and normal cytogenetics: a study of the Acute Myeloid Leukemia Study Group Ulm. J Clin Oncol 2002:20:3254–61. doi: 10.1200/JCO.2002.09.088. [DOI] [PubMed] [Google Scholar]
  • 112.Falini B, Brunetti L, Sportoletti P, and Martelli MP. NPM1-mutated acute myeloid leukemia: from bench to bedside. Blood 2020:136):1707–1721. 10.1182/blood.2019004226 [DOI] [PubMed] [Google Scholar]
  • 113.Brown P, McIntyre E, Rau R, Meshinchi S, Lacayo N, Dahl G, et al. The incidence and clinical significance of nucleophosmin mutations in childhood AML. Blood. 2007:110:979–985. doi: 10.1182/blood-2007-02-076604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Bhatlekar S, Fields JZ, and Boman BM. Role of HOX genes in stem cell differentiation and cancer. Stem Cells Int. 2018:3569493. doi: 10.1155/2018/3569493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Alharbi RA, Pettengell R, Pandha HS and Morgan R. The role of HOX genes in normal hematopoiesis and acute leukemia. Leukemia. 2013:27):1000–1008. doi: 10.1038/leu.2012.356. [DOI] [PubMed] [Google Scholar]
  • 116.Antunes ETB and Ottersbach K. The MLL/SET family and haematopoiesis. Biochim Biophys Acta Gene Regul Mech. 2020:1863:194579. doi: 10.1016/j.bbagrm.2020.194579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Milne TA, Briggs SD, Brock HW, Martin ME, Gibbs D, Allis CD, et al. MLL targets SET domain methyltransferase activity to Hox gene promoters. Mol Cell. 2002:10:1107–1117. doi: 10.1016/s10972765(02)00741-4. [DOI] [PubMed] [Google Scholar]
  • 118.Dorrance AM, Liu S, Yuan W, Becknell B, Arnoczky KJ, Guimond M, et al. Mll partial tandem duplication induces aberrant Hox expression in vivo via specific epigenetic alterations. J Clin Invest. 2006:116:2707–2716. doi: 10.1172/JCI25546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Yokoyama A, Wang Z, Wysocka J, Sanyal M, Aufiero DJ, Kitabayashi I, et al. Leukemia proto-oncoprotein MLL forms a SET1-like histone methyltransferase complex with menin to regulate Hox gene expression. Mol Cell Biol. 2004:24:5639–5649. doi: 10.1128/MCB.24.13.5639-5649.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Yokoyama A, Somervaille TC, Smith KS, Rozenblatt-Rosen O, Meyerson M, Cleary ML. The menin tumor suppressor protein is an essential oncogenic cofactor for MLL-associated leukemogenesis. Cell. 2005:123:207–218. doi: 10.1016/j.cell.2005.09.025. [DOI] [PubMed] [Google Scholar]
  • 121.Yokoyama A and Cleary ML. Menin critically links MLL proteins with LEDGF on cancer-associated target genes. Cancer Cell. 2008:14:36–46. doi: 10.1016/j.ccr.2008.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Chen YX, Yan J, Keeshan K, Tubbs AT, Wang H, Silva A, et al. The tumor suppressor menin regulates hematopoiesis and myeloid transformation by influencing Hox gene expression. Proc Natl Acad Sci U S A. 2006:103:1018–1023. doi: 10.1073/pnas.0510347103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Vassiliou GS, Cooper JL, Rad R, Li J, Rice S, Uren A, et al. Mutant nucleophosmin and cooperating pathways drive leukemia initiation and progression in mice. Nat Genet. 2011:43:470–475. doi: 10.1038/ng.796. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Dovey OM, Cooper JL, Mupo A, Grove CS, Lynn C, Conte N, et al. Molecular synergy underlies the co-occurrence patterns and phenotype of NPM1-mutant acute myeloid leukemia. Blood. 2017:130:1911–1922. doi: 10.1182/blood-2017-01-760595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Uckelmann HJ, Kim SM, Wong EM, Hatton C, Giovinazzo H, Gadrey JY, et al. Therapeutic targeting of preleukemia cells in a mouse model of NPM1 mutant acute myeloid leukemia. Science. 2020:367:586–590. doi: 10.1126/science.aax5863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Brunetti L, Gundry MC, Sorcini D, Guzman AG, Huang YH, Ramabadran R, et al. Mutant NPM1 Maintains the Leukemic State through HOX Expression. Cancer Cell. 2018:34:499–512. doi: 10.1016/j.ccell.2018.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Gu X, Ebrahem Q, Mahfouz RZ, Hasipek M, Enane F, Radivoyevitch T, et al. Leukemogenic nucleophosmin mutation disrupts the transcription factor hub that regulates granulomonocytic fates. J Clin Invest. 2018:128:4260–4279. doi: 10.1172/JCI97117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Kühn MW, Song E, Feng Z, Sinha A, Chen CW, Deshpande AJ, et al. Targeting chromatin regulators inhibits leukemogenic gene expression in NPM1 mutant leukemia. Cancer Discov. 2016:6:1166–1181. doi: 10.1158/2159-8290.CD-16-0237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Cierpicki T and Grembecka J. Challenges and opportunities in targeting the menin-MLL interaction. Future Med Chem. 2014:6:447–462. doi: 10.4155/fmc.13.214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Grembecka J, He S, Shi A, Purohit T, Muntean AG, Sorenson RJ, et al. Menin-MLL inhibitors reverse oncogenic activity of MLL fusion proteins in leukemia. Nat Chem Biol. 2012:8:277–284. doi: 10.1038/nchembio.773. •First pre-clinical report of a menin inhibitor
  • 131.Perner F and Armstrong SA. Targeting chromatin complexes in myeloid malignancies and beyond: From basic mechanisms to clinical innovation. Cells. 2020:9:2721. doi: 10.3390/cells9122721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.He S, Malik B, Borkin D, Miao H, Shukla S, Kempinska K, et al. Menin-MLL inhibitors block oncogenic transformation by MLL-fusion proteins in a fusion partner-independent manner. Leukemia. 2016:30:508–513. doi: 10.1038/leu.2015.144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Timmers HTM, ÖzyerliGöknar E and Nizamuddin S. A box of chemistry to inhibit the MEN1 tumor suppressor gene promoting leukemia. ChemMedChem. 2021. Doi: 10.1002/cmdc.202000972. Online ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Klossowski S, Miao H, Kempinska K, Wu T, Purohit T, Kim E, et al. Menin inhibitor MI-3454 induces remission in MLL1-rearranged and NPM1-mutated models of leukemia. J Clin Invest. 2020:130:981–997. doi: 10.1172/JCI129126. ••Report of an optimized menin inhibitor suitable for in vivo use
  • 135.Kessler L, Wu T, Grembecka J, Cierpicki T, Purohit T, Miaoand H, et al. Discovery of novel menin-MLL small molecule inhibitors that display high potency and selectivity in vitro and in vivo. European Journal of Cancer. 2016: 69S:88. 10.1016/S0959-8049(16)32859–3. [DOI] [Google Scholar]
  • 136.Wu T, Kessler L, Li S, Purohit T, Li S, Miao H, et al.A novel small molecule menin-MLL inhibitor for potential treatment of MLL-rearranged leukemias. Cancer Res 2017:77(13 Suppl):Abstract 5077. doi: 10.1158/1538-7445.AM2017-5077 [DOI] [Google Scholar]
  • 137.Burrows F, Wu T, Kessler L, Li S, Zhang J, Zarrinkar P, et al. A novel small molecule menin-MLL inhibitor for potential treatment of MLL-rearranged leukemias and NPM1/DNMT3A-mutant AML Mol Cancer Ther. 2018:17(1 Suppl):Abstract LB-A27. Doi: 10.1158/1535-7163.TARG-17-LB-A27. [DOI] [Google Scholar]
  • 138. Wang ES, Altman JK, Pettit K, De Botton S, Walter RP, Fenaux P, Burrows F, et al. Preliminary data on a phase 1/2A first in human study of the menin-KMT2A (MLL) inhibitor KO-539 in patients with relapsed or refractory acute myeloid leukemia. Blood. 2020:136(Supplement 1): 7–8. Doi: 10.1182/blood-2020-134942. ••First in man report of the menin inhibitor KO-539
  • 139.Libbrecht C, Xie HM, Kingsley MC, Haladyna JN, Riedel SS, Alikarami F, et al. Menin is necessary for long term maintenance of meningioma-1 driven leukemia. Leukemia. 2021. February 4. doi: 10.1038/s41375-021-01146-z. Online ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Dzama MM, Steiner M, Rausch J, Sasca D, Schönfeld J, Kunz K, et al. Synergistic targeting of FLT3 mutations in AML via combined menin-MLL and FLT3 inhibition. Blood. 2020:136:2442–2456. doi: 10.1182/blood.2020005037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Schmidt L, Heyes E, Scheiblecker L, Eder T, Volpe G, Frampton J, et al. CEBPA-mutated leukemia is sensitive to genetic and pharmacological targeting of the MLL1 complex. Leukemia. 2019:33:1608–1619. doi: 10.1038/s41375-019-0382-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Krivtsov AV, Evans K, Gadrey JY, Eschle BK, Hatton C, Uckelmann HJ, et al. A Menin-MLL Inhibitor Induces Specific Chromatin Changes and Eradicates Disease in Models of MLL-Rearranged Leukemia. Cancer Cell. 2019:36:660–673 doi: 10.1016/j.ccell.2019.11.001. ••``Report of a second, structurally distinct menin inhibitor
  • 143. McGeehan G. A first-in-class Menin-MLL1 antagonist for the treatment of MLL-r and NPM1 mutant leukemias. Presentation at the 2020 American Association for Cancer Research Virtual Annual Meeting I. April 27, 2020. ••First report of a menin inhibitor clinical trial
  • 144.Borthakur GM, Odenike O, Aldoss I, Rizzieri DA, Prebet T, Modi DA, et al. Biomarker modulation by mivebresib (ABBV-075) +/− venetoclax in relapsed/refractory acute myeloid leukemia. Blood 2019:134 (Supplement_1): 539 Doi: 10.1182/blood-2019-126854. [DOI] [Google Scholar]
  • 145.Mascarenhas J, Saab R, Brackman D, Modi DA, Abraham L, Ward JE et al. Two Phase 1b Studies Evaluating the Safety and Tolerability of BET Inhibitors, ABBV-744 and Mivebresib, as Monotherapies and in Combination with Ruxolitinib or Navitoclax in Patients with Myelofibrosis. Blood. 2020: 136(Supplement 1): 18–19. doi: 10.1182/blood-2020-137686 [DOI] [Google Scholar]
  • 146.Talpaz M, Rampal R, Verstovsek S, Harrison C, Drummond M, Kiladjian JJ, et al. CPI-0610, a bromodomain and extraterminal domain protein (BET) inhibitor, as monotherapy in advanced myelofibrosis patients refractory/intolerant to JAK inhibitor: update from phase 2 Manifest study. European Hematology Association 2020 Annual Meeting. Abstract EP1091. [Google Scholar]
  • 147.Mascarenhas J, Harrison C, Ptriarca A, Devos T, Palandri F, Rampal R, et al. CPI-0610, a bromodomain and extraterminal domain protein (BET) inhibitor, in combination with ruxolitinib, in JAK inhibitor treatment naïve myelofibrosis patients: update from the Manifest phase 2 study. European Hematology Association 2020 Annual Meeting. Abstract EP1084. [Google Scholar]
  • 148.Verstovsek S, Mascarenhas J, Kremyanskaya M, Hoffman R, Rampal R, Gupta V, et al. CPI-0610, bromodomain and extraterminal domain protein (BET) inhibitor, as ‘add-on’ to ruxolitinib (RUX), in advanced myelofibrosis patients with suboptimal response: update of manifest phase 2 study. European Hematology Association 2020 Annual Meeting. Abstract EP1083. [Google Scholar]
  • 149.Mascarenhas J, Harrison C, Luptakova K, Christo J, Wang J, Mertz JA, et al. MANIFEST-2, a global, phase 3, randomized, double-blind, active-control study of CPI-0610 and ruxolitinib vs. placebo and ruxolitinib in JAK-inhibitor-naive myelofibrosis patients. Blood. 2020:136 (Supplement 1): 43. Doi: 10.1182/blood-2020-140901 [DOI] [Google Scholar]
  • 150.Patel MR, Garcia-Manero G, Paquette R, Dinner S, Donnellan WB, Grunwald MR, et al. Phase 1 dose escalation and expansion study to determine safety, tolerability, pharmacokinetics, and pharmacodynamics of the BET inhibitor FT-1101 as a single agent in patients with relapsed or refractory hematologic malignancies Blood (2019) 134 (Supplement_1): 390 [Google Scholar]
  • 151.Dawson M, Stein EM, Huntly BJP, Karadimitris A, Kamdar M, de Larrea CF, et al. A Phase I study of GSK525762, a selective bromodomain (BRD) and extra terminal protein (BET) inhibitor: results from part 1 of phase I/II open label single agent study in patients with acute myeloid leukemia (AML). Blood 2017:130 (Supplement 1): 1377. Doi: 10.1182/blood.V130.Suppl_1.1377.1377.28667012 [DOI] [Google Scholar]
  • 152.Lihou C, Zhou G, and Zheng F. A phase 1 study of INCB057643 monotherapy in patients with relapsed or refractory myelofibrosis (INCB 57643–103). Blood 2020:136 (Supplement 1): 16–17. Doi: 10.1182/blood-2020-134604 [DOI] [Google Scholar]
  • 153.Mims AS, Solh MD, Borate U, Pemmaraju N, Borthakur G, Roboz GJ, et al. Phase 1b study of BET inhibitor PLX2853 in patients with relapsed or refractory acute myeloid leukemia or high risk myelodysplastic syndrome. Blood 2020:136 (Supplement 1):14–15. Doi: 10.1182/blood-2020-140138 [DOI] [Google Scholar]
  • 154.Patnaik A, Carvajal RD, Komatsubara KM, Britten CD, Wesolowski R, Michelson G, et al. Phase ib/2a study of PLX51107, a small molecule BET inhibitor, in subjects with advanced hematological malignancies and solid tumors. J Clin Oncol 2018:36: Abstract 2550. Doi: 10.1200/JCO.2018.36.15_suppl.2550. [DOI] [Google Scholar]
  • 155.Roboz GJ, Desai P, Lee S, Ritchie EK, Winer ES, DeMario M, et al. A dose escalation study of RO6870810/TEN-10 in patients with acute myeloid leukemia and myelodysplastic syndrome. Leuk Lymphoma 2021. February 13;1–13. doi: 10.1080/10428194.2021.1881509. Online ahead of print. [DOI] [PubMed] [Google Scholar]
  • 156.Murphy T, Leber B, Bray MR, Chan SM, Gupta V, Khalaf D, et al. Preliminary results from a phase 1 study of Cfi-400495, a PLK4 inhibitor, in patients with acute myeloid leukemia and high risk MDS. Blood 2020:136(Supplement 1):1–2. Doi: 10.1182/blood-2020-138822.32430499 [DOI] [Google Scholar]
  • 157.Jonas BA, Bixby DL, Brandwein JM, Yee KWL, Murphy T, Minden MD, et al. A phase 2 open-label, multicenter, dose optimization clinical study of the safety, tolerability, and pharmacokinetic (PK) and pharmacodynamic (PD) profiles of Cfi-400945 as a single agent or in combination with azacitidine or decitabine in patients with acute myeloid leukemia. Blood 2020:136 (Supplement 1):24–25. Doi: 10.1182/blood-2020-13600432430494 [DOI] [Google Scholar]
  • 158.Navada SC, Garcia-Manero G, OdchimarReissig R, Pemmaraju N, Alvarado Y, Ohanian MN, et al. Rigosertib in combination with azacitidine in patients with myelodysplastic syndromes or acute myeloid leukemia: results of a phase 1 study. Leuk Res 2020:94:106369. Doi: 10.1016/j.leukres.2020.106369. [DOI] [PubMed] [Google Scholar]
  • 159.Navada SC, Fruchtman SM, Odchimar-Reissig R, Demakos EP, Petrone ME, Zbyszewski PS, et al. A phase 1/2 study of rigosertib in patients with myelodysplastic syndromes (MDS) and MDS progressed to acute myeloid leukemia. Leuk Res 2018:64:10–16. Doi: 10.1016/j.leukres.2017.11.006. [DOI] [PubMed] [Google Scholar]
  • 160.Garcia-Manero G,Fenaux P, Al-Kali A, Baer MR, Sekeres MA, Roboz GJ, et al. Rigosertib versus best supportive care for patients with high-risk myelodysplastic syndromes after failure of hypomethylating drugs (ONTIME): a randomized, controlled, phase 3 trial. Lancet Oncol 2016: 17:496–508. Doi: 10.1016/S1470-2045(16)00009-7. [DOI] [PubMed] [Google Scholar]

RESOURCES