Targeting menin in relapsed or refractory acute myeloid leukaemia—where the rubber hits the road
Rearrangements of the lysine methyltransferase 2A [KMT2A, formerly known as mixed lineage leukaemia (MLL)], chromosome locus 11q23—initially established as target for leukaemia therapy by Yokoyama et al. are rare alterations and occur in up to 10% of all acute leukaemias (1). Moreover, it is often associated with pleiotropic drug resistance and high rates of relapse (2). The medium progression-free survival (mPFS) has been reported to be 30–40% with a medium overall survival (mOS) of less than 25% (3). Acute myeloid leukaemias (AMLs) harbouring a KMT2A rearrangement are known to have an aggressive outcome and are associated with early relapse, massive hyperleukocytosis, high incidence of involvement of the central nervous system involvement in most cases, and a very dismal prognosis. In addition, in adults KMT2A rearrangements are typically found in patients with prior treatments with topoisomerase-II (TOPO-II) inhibitors (e.g., etoposide, teniposide) (4). KMT2A rearrangements (with more than 80 fusion partners having been identified so far) have been found in 5–10% of adult AML patients and are very common in infant leukaemia (70–80%) (5).
KMT2A acts as a significant epigenetic gene regulator and plays a major role for the haematopoiesis. In addition, KMT2A is the master regulator for key genes such as HOXA and HOXB, MEIS1, PBX3, MEF2C and CDK6 (6) and thereby promotes self-renewal of myeloid progenitor cells. Of note, KMT2A rearrangements have not only been detected in AML patients, but also can be detected in acute lymphoblastic leukaemia (ALL) and in mixed-phenotype acute leukaemia (MPAL) (7).
Menin is a nuclear scaffold protein which is involved in various biological pathways, and interacts with the N-terminal part of KMT2A, and the menin-KMT2A interaction is required for KMT2A-fusion target gene expression, such as HOXA and MEIS1, which are essential drivers of KMT2A-fusion leukaemogenesis (Figure 1) [reviewed by (10)]. Several preclinical and clinical research papers have provided significant evidence that highly specific menin inhibitors can cause downregulation of the KMT2A-fusion targets such as HOX, PBX3, and MEIS1 genes, which then leads to differentiation of leukaemic blasts (6,11).

Nucleophosmin 1 (NPM1) is a chaperone protein and its mutated form serves as a shuttle between the nucleus and the cytoplasm (8). NPM1 mutations frequently co-occur with other genetic alterations, mainly Flt-3 internal tandem duplications (Flt3-ITD) (60%) and DNMT3A mutations (50%) (8,11). Moreover, NPM1 mutations in AML are known to be associated with a favourable prognosis, particularly without concurrent high-risk mutations such as FLT3-ITD. Several clinical trials have provided compelling evidence that AML patients harbouring NPM1 mutations have improved complete response (CR) rates and longer mOS compared to those patients without these mutations (11).
Interestingly, in AML cells harbouring NPM1 mutations (found approximately in 30% of AML patients), rearranged KMT2A is the key oncogenic driver of HOX, MEIS1, and Flt-3 (12). Of note, NPM1 has been shown to bind to specific chromatin targets, which are co-occupied by KMT2A, and menin inhibition has been shown to initiate degradation of mNPM1 resulting in a rapid decrease in gene expression and activating histone modifications (8,12,13) which adds weight to the proposal that mutations of NPM1 are essential targets for the menin-KMT2A inhibition in leukaemias. This important observation adds weight to the proposal that blockage of the menin-KMT2A axis inhibits leukaemias driven by NPM1 mutations for which the expression of KMT2A target genes is essential (14). It should be noted that preclinical results also suggest that the KMT2A inhibition by menin inhibitors may be a novel therapy for AML patients harbouring rearrangements of the nucleoporin 98 gene (NUP98) (mainly associated with infant leukaemias) since NUP98-fusion-driven AML cells were found to be sensitive to menin inhibitors in experimental systems (15,16).
These potential clinical findings prompted several pharmaceutical companies to search for inhibitors targeting the NPM1 mutations in AML which quickly led to the development of novel, oral, highly potent, and specific menin inhibitors (Table 1). All currently available small molecule menin inhibitors are innovative targeted agents currently under clinical evaluation for AML patients harbouring KMT2A and NUP98 rearrangements or NPM1 mutations (17,18).
Table 1
Compound (manufacturer) | Trial | Phase | Regimen | Comments |
---|---|---|---|---|
Revumenib (Syndax) | NCT04065399 (AUGMENT-001) | I/II | Monotherapy | r/r AML (KMP2A and mNPM1) |
(ORR 68.2%, CR/CRh rate: 22.8%) | ||||
NCT05326516 (AUGMENT-102) | I | Revumenib/FLA (AML) | r/r AML (KMP2A and mNPM1) or NUP98r | |
Revumenib/Pred/VCR/ASP/DNR (ALL) | (CR rate: 52%) | |||
NCT05761171 | II | Revumenib/FLA/MTX | r/r AML (KMT2A) or ALL | |
NCT05360160 (SAVE) | I/II | Revumenib + ASTX727/VEN | r/r AML or ALL | |
(ORR 100%, CR rate: 33%) | ||||
NCT030113998 (BEAT) | I | Revumenib + VEN/AZA (AML) | Newly diagnosed AML (KMP2A and mNPM1) (CR rate: 96%) |
|
Ziftomenib (Kura Oncology) | NCT04067336 (KOMET-001) | I/II | Monotherapy | Phase I: r/r AML |
mNPM1: ORR 40%, CR rate: 30% | ||||
NCT05735184 (KOMET-007) | I | Ziftomenib plus 7 + 3 (AML) | KMT2A: ORR 16.7%, CR rate: 11% | |
Ziftomenib plus VEN/AZA (r/r AML) | Phase I: AML or r/r AML | |||
First-line: CR 100% | ||||
Second-line: ORR 53% (CRh rate mNPM1: 60%, CRh rate KMT2A: 50%) | ||||
Bleximenib (Johnson & Johnson) | NCT04811560 | I | Monotherapy | r/r AML (KMP2A and mNPM1) |
NCT05453903 | I | Bleximenib plus VEN or AZA or both | r/r AML (KMP2A and mNPM1) | |
Bleximenib plus FLA (AML) | ||||
NCT05521087 | I | Bleximenib plus DEX/VCR/ASP (ALL) | r/r AML (KMP2A and mNPM1) | |
BMF-219 (BioMea) | NCT05153301 (COVALENT-101) | I | Monotherapy | r/r AML, DLBCL, or MM (CR rate: 2/5 patients) |
Emilumenib (Daiichi-Sankyo) | NCT04752163 | I/II | Emilumenib ± AZA, VEN, or mini-HCVD | Phase I: r/r AML |
Phase II: r/r AML/ALL (KMT2A), AML (NPM1) | ||||
DSP-5336 (Sumitomo Dainippon Pharma) | NCT04988555 | I/II | Monotherapy | Phase I: r/r AML |
Phase II: r/r AML/ALL (KMT2A), AML (NPM1) (ORR 57%, CR rate: 24%) | ||||
BN-104 (Yehui Pharma) | NCT060528133 | I/II | Monotherapy | r/r AML (KMP2A and mNPM1) |
Of note, BMF-219 is the only menin inhibitor that also targets the MYC oncogene. r/r AML, relapsed/refractory acute myeloid leukaemia; ORR, overall response rate; CR, complete response; CRh, complete remission with partial hematological recovery; FLA, fludarabine plus cytarabine; Pred, prednisone; VCR, vincristine; ASP, PEG-asparaginase; DNR, daunorubicin; ALL, acute lymphoblastic leukaemia; MTX, methotrexate; ASTX727, cedazuridin/decitabine; VEN, venetoclax; AZA, azacitidine; DEX, dexamethasone; DLBCL, diffuse large B cell lymphoma; MM, multiple myeloma.
Most recently, Issa et al. (19) have reported the results of the AUGMENT-101 trial for revumenib in r/r AML patients harbouring KMT2A rearrangements or NPM1 mutations (phase I/II trial, NCT04065399). The primary endpoints in this study were the CR/CRh (complete response with partial hematologic recovery) rate and safety. Revumenib was administered at a dose of 163 mg BID until progression or unacceptable toxicities. Patients enrolled were heavily pretreated, with approximately 44% having received three or more treatment lines including stem cell transplants (median 2, range 1–11). Amongst the 94 patients enrolled, grade ≥3 adverse events were found to be febrile neutropenia (37.2%), differentiation syndrome (16%), and QTc prolongation (13.8%). Revumenib was well tolerated and the observed differentiation syndrome was manageable with steroids and hydroxyurea. A total of 57 patients were eligible for the efficacy evaluation, and the CR/CRh rate was reported to be 22.8% with an overall response rate (ORR) of 63.2%. Of note, 15 of 22 patients (68.2%) had no detectable minimal residual disease (MRD). Interestingly, responses were found to occur rapidly (median of 0.95 months). Since the KMT2A rearrangement is a rare subtype, the trial was conducted as a single-arm study. Moreover, a mutation analysis was not performed following progression, therefore, the underlying resistance mechanisms remain unclear.
Although the mNPM1 part of the trial is still ongoing, the authors concluded that treatment with revumenib has a favourable toxicity profile and is highly active in r/r AML patients harbouring KMT2A rearrangements suggesting that revumenib warrants further evaluation in newly diagnosed AML patients.
In the AUGMENT-101 phase I trial, the most common adverse events included QTc prolongation (25.5% any grade, 13.8% ≥ grade 3), vomiting (30.9%), nausea (44.7% any grade) and febrile neutropenia (38.3% any grade, 37.2% ≥ grade 3). Of note, the differentiation syndrome is a treatment-related event that is seen in almost all clinical trials with menin. The underlying mechanisms are supposed to be cytokine alterations associated with haematopoietic differentiation and can include fever, arthralgias, leukocytosis, pleural or pericardial effusions, and respiratory or renal failure in severe cases (7). It has been reported in 16% of patients in the AUGMENT-101 trial and in 21% (all grades) of ziftomenib-treated patients (KOMET-001, N=83) (20), however, it appeared to be clinically manageable in the AUGMENT-101 trial and did not require dose modifications (19). Based on the results form the AUGMENT-101 trial, FDA approved remuvenib (Revuforj®) for treatment of r/r AML patients harbouring KMP2A rearrnagements on November 15, 2024.
During the last four to five decades the identified underlying molecular alterations seen in AML patients had not demonstrated any significant impact on the first-line induction chemotherapy. The vast majority clinicians still treat AML patients who are eligible for intensive chemotherapy with an anthracycline (e.g., daunorubicin or idarubicin for 3 consecutive days) plus ara-C (continuous infusion over 7 days) (generally known as the “7 + 3” regimen) (8). A significant breakthrough in terms of mOS benefit was recently demonstrated by a new “7 + 3” formulation (CPX-351, Vyxeos liposomal®) in patients with secondary AML (21). Until now, several mutation-specific targeted agents such as inhibitors of Flt-3, isocitrate dehydrogenase (IDH1,2), bcl-2, and hedgehog have been approved by FDA and European Medicines Agency (EMA) [reviewed by (11)]. However, despite these advances, it still remains a challenge to treat certain subgroups of AML patients.
The current clinical development of menin inhibitors strongly supports the ongoing paradigm shift towards targeted therapies for treatment of AML patients. These drugs appear to be very innovative molecules and represent a new class of attractive inhibitors of AML driver mutations. Although first results with menin inhibitors used as single agents (Table 1) appear to be very promising, they tend to be insufficient for achieving long and robust improvements in clinical outcome. Early data clearly indicated that AML patients who initially responded to menin inhibitors but then relapsed, were found to have new alterations in the menin gene suggesting that current inhibitors such as revumenib may no longer disrupt menin from binding to KMT2A (9,22). It is conceivable, therefore, that the development of both intrinsic and acquired resistance to menin inhibitors may limit their therapeutic activity, and some researches have provided evidence that up to 40% of these resistant patients are associated with menin gene mutations, whereas the menin gene was found to be unaltered in other studies suggesting that non-mutational resistance mechanisms may also be involved (22,23). Moreover, little is known about the development of resistant clones in terms of the rapid acquisition of treatment failures versus the observed sustainable responses seen in some studies (median duration of 9.1 months) (10).
The majority of clinical studies so far has been conducted in the r/r AML setting (Table 1). More than 22,000 r/r AML patients are diagnosed in Europe every year with an estimated median survival of less than three months (Prof. Giovanni Martinelli, Bologna, Italy, personal communication). For those patients who are not eligible for intensive salvage therapy, very few approved and experimental treatment options with low CR rates (approximately 20–30%) are available (Table 2) indicative of a high unmet medical need for this cohort. Reported CR rates from initial phase I/II trials with menin inhibitors suggest a better treatment outcome with these compounds, however, it is too early to draw definitive conclusions and only a subgroup of r/r AML patients may be selected for treatment. In this regard, an innovative non-cardiotoxic anthracycline derivative (L-annamycin) (24,25) has sparked considerable interest as a combination partner for menin inhibitors.
Table 2
Compound | Manufacturer | Target | Regimen | Outcome |
---|---|---|---|---|
Gilteritinib | Astellas (Japan) | Flt-3 | Monotherapy | CR rate 34%; mOS 9.3 months (approved for r/r AML patients with Flt-3 alterations after AZA/VEN) |
Gilteritinib + venetoclax | Astellas (Japan) & Abbvie (USA) | Flt-3, bcl-2 | Combination | CR rate 18%, mOS 10 months (phase I) |
Phase II/III in preparation | ||||
Venetoclax plus azacitidine plus gilteritinib | Astellas (Japan), Celgene (USA) & AbbVie (USA) | bcl-2, HMA, Flt-3 | Combination | CR rate 18%, mOS 5.8 months |
Venetoclax plus decitabine/ceda-zuridine | Taiho (Japan), AbbVie (USA) | bcl-2, HMA, cytidine deaminase inhibitor | Combination | Results are pending |
MGD024 | MacroGeneics (USA) | CD123 × CD3 | Monotherapy | Study ongoing (NCT05362773) |
Debio 1562M | Debiopharm (CH) | CD37 | Monotherapy | Study in preparation |
Olutasidenib | Rigel Pharmaceuticals (USA) | IDH-1 | Monotherapy | ORR 48%, CR rate: 32%, mOS 11.6 months (approved for r/r AML patients harbouring IDH-1 mutations) |
Enasidenib | Celgene (USA) | IDH-2 | Monotherapy | ORR 40.3%, CR rate 19.3%, mOS 9.3 months (19.3 months for patients with CR) |
L-Annamycin | Moleculin (USA) | TOPO-I/II | L-annamycin + araC | MB-106: CR rate 60% |
MB-108 (MIRACLE): study in preparation |
r/r AML, relapsed/refractory acute myeloid leukaemia; Flt-3, Fms-like tyrosine kinase 3; CR, complete response; mOS, medium overall survival; AZA, azacitidine; VEN, venetoclax; HMA, hypomethylating agent; CD, cluster of differentiation; IDH, isocitrate dehydrogenase; ORR, overall response rate; TOPO, topoisomerase; araC, cytarabine.
It is still too early to answer the question how to use menin inhibitors and how to incorporate them into a comprehensive therapeutic strategy. In addition, it is far from being clear whether menin inhibitors should be used as part of large combinations treatments or successions of treatments.
Trials combining menin inhibitors with standard induction chemotherapy or venetoclax-based regimens for both newly diagnosed and r/r AML patients are currently open for recruitment, and final results from these studies are eagerly awaited. Since menin inhibitors appear to have a synergistic effect with many other agents (Flt-3 inhibitors, hypomethylating agents, venetoclax, novel cytostatic drugs etc.), it is conceivable that the combination of both classes of drugs may improve outcomes of AML patients harbouring KMT2A or NPM1 alterations, especially in older patients and in those with r/r AML, and thereby pave the way for better AML therapies in the near future.
Acknowledgments
None.
Footnote
Provenance and Peer Review: This article was commissioned by the editorial office, AME Clinical Trials Review. The article has undergone external peer review.
Peer Review File: Available at https://actr.amegroups.com/article/view/10.21037/actr-24-250/prf
Funding: None.
Conflicts of Interest: Both authors have completed the ICMJE uniform disclosure form (available at https://actr.amegroups.com/article/view/10.21037/actr-24-250/coif). The authors have no conflicts of interest to declare.
Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.
Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.
References
- Yokoyama A, Somervaille TC, Smith KS, et al. The menin tumor suppressor protein is an essential oncogenic cofactor for MLL-associated leukemogenesis. Cell 2005;123:207-18. [Crossref] [PubMed]
- Issa GC, Aldoss I, DiPersio J, et al. The menin inhibitor revumenib in KMT2A-rearranged or NPM1-mutant leukemia. Nature 2023;615:920-4. [Crossref] [PubMed]
- Guarnera L, D'Addona M, Bravo-Perez C, et al. KMT2A Rearrangements in Leukemias: Molecular Aspects and Therapeutic Perspectives. Int J Mol Sci 2024;25:9023. [Crossref] [PubMed]
- Issa GC, Zarka J, Sasaki K, et al. Predictors of outcomes in adults with acute myeloid leukemia and KMT2A rearrangements. Blood Cancer J 2021;11:162. [Crossref] [PubMed]
- Pendleton M, Lindsey RH Jr, Felix CA, et al. Topoisomerase II and leukemia. Ann N Y Acad Sci 2014;1310:98-110. [Crossref] [PubMed]
- Swaminathan M, Bourgeois W, Armstrong SA, et al. Menin Inhibitors in Acute Myeloid Leukemia-What Does the Future Hold? Cancer J 2022;28:62-6. [Crossref] [PubMed]
- Issa GC, Ravandi F, DiNardo CD, et al. Therapeutic implications of menin inhibition in acute leukemias. Leukemia 2021;35:2482-95. [Crossref] [PubMed]
- Patel SS. NPM1-Mutated Acute Myeloid Leukemia: Recent Developments and Open Questions. Pathobiology 2024;91:18-29. [Crossref] [PubMed]
- Perner F, Stein EM, Wenige DV, et al. MEN1 mutations mediate clincial resistance to menin inhibitors. Nature 2023;615:913-9. [Crossref] [PubMed]
- Thomas X. Small Molecule Menin Inhibitors: Novel Therapeutic Agents Targeting Acute Myeloid Leukemia with KMT2A Rearrangement or NPM1 Mutation. Oncol Ther 2024;12:57-72. [Crossref] [PubMed]
- Nong T, Mehra S, Taylor J. Common Driver Mutations in AML: Biological Impact, Clinical Considerations, and Treatment Strategies. Cells 2024;13:1392. [Crossref] [PubMed]
- Kühn MWM, Ganser A. The Menin story in acute myeloid leukaemia-The road to success. Br J Haematol 2024;205:812-4. [Crossref] [PubMed]
- Falini B, Mecucci C, Tiacci E, et al. Cytoplasmatic nucleophosmin in acute myelogenous leukemia with a normal karyotype. N Engl J Med 2005;352:254-66. [Crossref] [PubMed]
- Dempke WCM, Desole M, Chiusolo P, et al. Targeting the undruggable: menin inhibitors ante portas. J Cancer Res Clin Oncol 2023;149:9451-9. [Crossref] [PubMed]
- Fiskus W, Boettcher S, Daver N, et al. Effective menin inhibitor-based combination against AML with MLL rearrangement or NPM1 mutation (NPM1c). Blood Cancer J 2022;12:5. [Crossref] [PubMed]
- Uckelmann HJ, Haarer EL, Takeda R, et al. Mutant NPM1 Directly Regulates Oncogenic Transcription in Acute Myeloid Leukemia. Cancer Discov 2023;13:746-65. [Crossref] [PubMed]
- Ranieri R, Pianigiani G, Sciabolacci S, et al. Current status and future perspectives in targeted therapy of NPM1-mutant AML. Leukemia 2022;36:2351-67. [Crossref] [PubMed]
- Heikamp EB, Henrich JA, Perner F, et al. The menin-MLL1 interaction is a molecular dependency in NUP98-rearranged AML. Blood 2022;139:894-906. [Crossref] [PubMed]
- Issa GC, Aldoss I, Thirman MJ, et al. Menin Inhibition With Revumenib for KMT2A-Rearranged Relapsed or Refractory Acute Leukemia (AUGMENT-101). J Clin Oncol 2025;43:75-84. [Crossref] [PubMed]
- Wang ES, Issa GC, Erba HP, et al. Ziftomenib in relapsed or refractory acute myeloid leukaemia (KOMET-001): a multicentre, open-label, multi-cohort, phase 1 trial. Lancet Oncol 2024;25:1310-24. [Crossref] [PubMed]
- Lancet JE, Uy GL, Cortes JE, et al. CPX-351 (cytarabine and daunorubicin) Liposome for Injection Versus Conventional Cytarabine Plus Daunorubicin in Older Patients With Newly Diagnosed Secondary Acute Myeloid Leukemia. J Clin Oncol 2018;36:2684-92. [Crossref] [PubMed]
- Zhou X, Zhang L, Aryal S, et al. Decoding the epigenetic drivers of menin-MLL inhibitor resistance in KMT2A-rearranged acute myeloid leukemia. ASH Meeting 2023;abstract 587.
- Candoni A, Coppola G A. 2024 Update on Menin Inhibitors. A New Class of Target Agents against KMT2A-Rearranged and NPM1-Mutated Acute Myeloid Leukemia. Hematol Rep 2024;16:244-54. [Crossref] [PubMed]
- Dempke WCM, Zielinski R, Winkler C, et al. Anthracycline-induced cardiotoxicity - are we about to clear this hurdle? Eur J Cancer 2023;185:94-104. [Crossref] [PubMed]
- Gil L, Wierzbowska A, Wrobel T, et al. Results of a phase I study of liposomal annamycin for treatment of r/r AML patients after induction therapy. Br J Cancer Res 2023;6:613-22.
Cite this article as: Dempke WCM, Fenchel K. Targeting menin in relapsed or refractory acute myeloid leukaemia—where the rubber hits the road. AME Clin Trials Rev 2025;3:7.