Revumenib: a new potential targeted therapy for KMT2A-rearranged acute leukemia
Editorial Commentary

Revumenib: a new potential targeted therapy for KMT2A-rearranged acute leukemia

Xavier Thomas

Department of Clinical Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre-Bénite, France

Correspondence to: Xavier Thomas, MD. Department of Clinical Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, 165 Chemin du Grand Revoyet, 69495 Pierre-Bénite Cedex, France. Email: xavier12thomas@gmail.com.

Comment on: 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.


Keywords: Menin inhibitors; acute leukemias; revumenib; KMT2A; targeted therapy


Received: 18 September 2024; Accepted: 20 December 2024; Published online: 28 February 2025.

doi: 10.21037/actr-24-188


Despite major therapeutic advances over the last decades, acute leukemia still represents an aggressive disease with a poor prognosis, especially for patients who are either considered unfit for intensive treatment or with relapsed or refractory disease. Thus relapsed/refractory acute leukemia remains an unsolved therapeutic problem, for which therapies with alternative mechanism of action are needed.

Thanks to a better understanding of mechanisms governing leukemia biology, the recent years have seen the development of many trials investigating targeted drugs, of which some have shown promising results able to change the standard of care in the treatment of patients with high-risk leukemia. In this setting, several mutation-specific targeted agents have been recently approved by the US and/or the European health agencies. These novel agents include Fms-like tyrosine kinase 3 (FLT3) inhibitors (1,2), isocitrate dehydrogenase (IDH) inhibitors (3,4), and venetoclax, a B cell leukemia/lymphoma 2 (BCL-2) inhibitor (5,6).

Menin inhibitors are novel targeted agents in acute leukemias. They target acute leukemias with rearrangement of the lysine methyltransferase 2A (KMT2A) gene (previously known as mixed-lineage leukemia) and those with mutation of the nucleophosmin 1 (NPM1) gene (7). Menin acts as a scaffolding protein/adaptor protein, providing a platform for other proteins to interact and regulate gene expression, particularly in the context of HOX genes. Menin binds to KMT2A, a histone methyltransferase, which is critical for the proper methylation of histone marks associated with HOX gene activation. Regulation of gene expression involves interaction with various transcription factors and chromatin regulators. The binding site is a cofactor for binding to HOX gene promoters. The overexpression of HOX genes and their cofactor MEIS1 participates to leukemic transformation through a blockage of differentiation (8). While NPM1 gene mutations are often considered as having a favorable prognosis, rearrangements of KMT2A, common in infants and children with acute leukemia, are especially associated with poor outcomes after conventional chemotherapy. They occur in up to 10% of acute leukemias (rearrangements of KMT2A occur in 80% of infant acute lymphoblastic leukemia and 5–15% of children and adults with acute myeloid, lymphoblastic or mixed phenotype leukemia) and definitively require the development of alternative therapies (9). Until now no targeted therapies were approved for these unfavorable molecular abnormalities.

In a recent issue of the Journal of Clinical Oncology, Issa and colleagues (10) reported findings from a phase 2 study of 94 patients, with relapsed or refractory KMT2A-rearranged acute leukemia of any lineage, given revumenib, an oral small molecule inhibitor of the menin-KMT2A interaction, administered as monotherapy (163 mg once every 12 hours, with a strong cytochrome P450 inhibitor, in 28-day cycles). Revumenib led to high remission rates with an overall response rate of 63.2% and a complete remission (CR) plus CR with partial hematologic recovery (CRh) rate of 22.8% (24.5% in acute myeloid leukemia and 14.3% in acute lymphoblastic leukemia) that exceeded the null hypothesis of expected remission rates for this patient population, even in heavily pretreated patients. Measurable residual disease (MRD)-negative response was observed in 70% of evaluable patients with CR plus CRh. The median duration of CR plus CRh was 6.4 months and the median overall survival was 8 months. The safety profile was acceptable showing grade ≥3 adverse events in 86% including essentially febrile neutropenia (37.2%), differentiation syndrome (16%), and QTc prolongation (13.8%) that generally resolved with dose adjustment and without treatment discontinuation. Overall, this paper confirmed the importance of the menin inhibitor revumenib in this molecularly defined sub-population of patients with relapsed/refractory acute leukemia.

This study demonstrates that menin inhibitor revumenib as monotherapy can provide clinical benefit in KMT2A-rearranged acute leukemias. Following these encouraging results, some issues should be mentioned.

Results should be interpreted with caution considering the relatively small number of patients included in the study, the short follow-up period, and the lack of control group although as mentioned by the authors this was not feasible due to the low frequency of the rearranged KMT2A leukemia subtype.

Although efficacy was noted with revumenib as monotherapy, a major issue concerns a potential better efficacy in combination with other available treatments. The good tolerance profile of revumenib pleads for the use of such therapeutic strategies, which might also reduce the risk of resistant clonal outgrowth. Furthermore, combinations should lead to deeper responses, and increased uptake of allogeneic stem cell transplantation (SCT), which remains the main therapeutic goal in relapsed/refractory patients. An emerging point was therefore the feasibility of SCT after revumenib therapy. In Issa et al.’ study (10), 38.9% of patients achieving response could proceed to allogeneic SCT, of which half followed post-transplant maintenance therapy with revumenib. In fit patients, studies incorporating revumenib into conventional intensive induction and consolidation chemotherapy backbones are currently ongoing (NCT05886049, NCT05761171). In unfit patients, preliminary data suggest high response rates when combining revumenib with venetoclax and a hypomethylating agent (NCT05360160) (11). Studies are also investigating combinations with other inhibitors, such as FLT3 inhibitors (gilteritinib, midostaurin) (NCT06222580, NCT06313437). All these studies should demonstrate feasibility and increased treatment efficacy. Question then will remain about best to use the combination therapy as part of a large combination or succession of treatments.

A notable treatment-related adverse event observed with revumenib therapy was differentiation syndrome, which is a common side effect observed with low-intensity targeted therapy. Symptoms are similar to those previously observed after other low-intensity treatments such as IDH inhibitors (12). This syndrome is mainly due to various cytokine alterations. Detection of differentiation syndrome justifies a successful reversal of the differentiation block. In Issa et al.’ study, differentiation syndrome was successfully managed with corticosteroids and hydroxyurea in case of leukocytosis. There were no treatment discontinuations (10).

In addition to revumenib, several other menin inhibitors are currently under investigation as monotherapy in a relapsed/refractory setting: KO-539 (ziftomenib) (NCT04067336), JNJ-75276617 (NCT04811560), DSP-5336 (NCT04988555), BMF-219 (NCT05153330), BN104 (NCT06052813) (13). Some of those agents are also currently tested in combination with other targeted agents or low-intensity therapy in unfit patients, or with intensive chemotherapy in patients considered fit: JNJ-75276617 with venetoclax and azacitidine (NCT05453903), ziftomenib with venetoclax, azacitidine, intensive chemotherapy, low-dose cytarabine, or gilteritinib (NCT05735184), DS-1594b with venetoclax and azacitidine (NCT04752163). All menin inhibitors should demonstrate efficacy in acute leukemias sharing the same dependency on the menin-KMT2A interaction. This includes leukemias displaying the NPM1 mutation and other less common subtypes (14,15). They could therefore have a larger application since acute myeloid leukemia with NPM1 mutations are seen in up to 40% of cases. Furthermore, preliminary results with revumenib are very encouraging in this setting (21% CR/CRh) and final findings should confirm those observed in KMT2A-rearranged leukemia (8).

Finally, as for all new agents, a major question is to understand whether revumenib has an effect on leukemia stem cells (LSCs). LSCs play a key role in the development and maintenance of acute leukemia (16). Quiescent leukemia-initiating cells escape to cytotoxic cancer agents which target rapidly proliferating cells, contributing then to treatment failure. Strategies for targeting LSCs hold then the promise to identify a more effective therapeutic approach to cure acute leukemia. Genes involved in self-renewal might represent rational targets for therapeutic intervention. Accumulating evidence suggests that KMT2A-rearranged leukemias originate from an uncommitted precursor (17). Stem cell-like blasts have been demonstrated in KMT2A-rearranged leukemias (18) and stem cell factor have been involved in KMT2A-rearranged leukemogenesis (19). A common theme in the various forms of KMT2A rearrangements is that KMT2A fusion proteins aberrantly upregulate the expression of HOX genes. Those ones play an essential role in the regulation of hematopoiesis and are highly expressed in hematopoietic stem cells and early progenitors (20). Their overexpression was shown to be sufficient for the initiation of leukemic transformation (21). Furthermore, up to 15% of patients with mixed phenotype acute leukemia (MPAL), which are position in the spectrum of immature leukemias, have rearrangements of KMT2A (22). In Issa et al.’s study (10), decreased expressions of MEIS1, HOXA9, PBX3, and FLT3 were noted. These findings are consistent with a potential action on LSCs. However, this decreased gene expression pattern was observed in both responders and non-responders to revumenib therapy and warrants confirmation in larger studies.

Much improvement is still needed in the treatment of high-risk patients with acute leukemia. Many drugs have become available that have the potential to change the standard of care for patients with acute leukemia. Because of their specific mechanism of action and their acceptable tolerance, menin inhibitors might be one of those effective new therapeutic strategies. They should rapidly be approved by health authorities for the treatment of leukemias with KMT2A-rearrangement or NPM1 mutation. Further inclusion into schedules combining several therapeutic agents might hold the promise of substantial benefit.


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-24-188/prf

Funding: None.

Conflicts of Interest: The author has completed the ICMJE uniform disclosure form (available at https://actr.amegroups.com/article/view/10.21037/actr-24-188/coif). The author has no conflicts of interest to declare.

Ethical Statement: The author is 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

  1. Stone RM, Mandrekar SJ, Sanford BL, et al. Midostaurin plus Chemotherapy for Acute Myeloid Leukemia with a FLT3 Mutation. N Engl J Med 2017;377:454-64. [Crossref] [PubMed]
  2. Perl AE, Altman JK, Cortes J, et al. Selective inhibition of FLT3 by gilteritinib in relapsed or refractory acute myeloid leukaemia: a multicentre, first-in-human, open-label, phase 1-2 study. Lancet Oncol 2017;18:1061-75. Erratum in: Lancet Oncol 2017;18:e711. [Crossref] [PubMed]
  3. Stein EM, DiNardo CD, Fathi AT, et al. Molecular remission and response patterns in patients with mutant-IDH2 acute myeloid leukemia treated with enasidenib. Blood 2019;133:676-87. [Crossref] [PubMed]
  4. DiNardo CD, Stein EM, de Botton S, et al. Durable Remissions with Ivosidenib in IDH1-Mutated Relapsed or Refractory AML. N Engl J Med 2018;378:2386-98. [Crossref] [PubMed]
  5. Wei AH, Strickland SA Jr, Hou JZ, et al. Venetoclax Combined With Low-Dose Cytarabine for Previously Untreated Patients With Acute Myeloid Leukemia: Results From a Phase Ib/II Study. J Clin Oncol 2019;37:1277-84. [Crossref] [PubMed]
  6. DiNardo CD, Pratz K, Pullarkat V, et al. Venetoclax combined with decitabine or azacitidine in treatment-naive, elderly patients with acute myeloid leukemia. Blood 2019;133:7-17. [Crossref] [PubMed]
  7. Issa GC, Ravandi F, DiNardo CD, et al. Therapeutic implications of menin inhibition in acute leukemias. Leukemia 2021;35:2482-95. [Crossref] [PubMed]
  8. Issa GC, Aldoss I, DiPersio J, et al. The menin inhibitor revumenib in KMT2A-rearranged or NPM1-mutant leukaemia. Nature 2023;615:920-4. [Crossref] [PubMed]
  9. 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]
  10. 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]
  11. Issa GC, Cuglievan B, DiNardo CD, et al. Early results of the phase I/II study investigating the all-oral combination of the menin inhibitor revumenib (SNDX-5613) with decitabine/cedazuridine (ASTX727) and venetoclax in acute myeloid leukemia (SAVE). Blood 2023;142:58. [Crossref]
  12. Fathi AT, Stein EM, DiNardo CD, et al. Differentiation syndrome with lower-intensity treatments for acute myeloid leukemia. Am J Hematol 2021;96:735-46. [Crossref] [PubMed]
  13. 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]
  14. Barajas JM, Rasouli M, Umeda M, et al. Acute myeloid leukemias with UBTF tandem duplications are sensitive to menin inhibitors. Blood 2024;143:619-30. [Crossref] [PubMed]
  15. Rasouli M, Blair H, Troester S, et al. The MLL-Menin Interaction is a Therapeutic Vulnerability in NUP98-rearranged AML. Hemasphere 2023;7:e935. [Crossref] [PubMed]
  16. Thomas D, Majeti R. Biology and relevance of human acute myeloid leukemia stem cells. Blood 2017;129:1577-85. [Crossref] [PubMed]
  17. Winters AC, Bernt KM. MLL-Rearranged Leukemias-An Update on Science and Clinical Approaches. Front Pediatr 2017;5:4. [Crossref] [PubMed]
  18. Chen C, Yu W, Alikarami F, et al. Single-cell multiomics reveals increased plasticity, resistant populations, and stem-cell-like blasts in KMT2A-rearranged leukemia. Blood 2022;139:2198-211. [Crossref] [PubMed]
  19. Yang L, Liu L, Gao H, et al. The stem cell factor SALL4 is an essential transcriptional regulator in mixed lineage leukemia-rearranged leukemogenesis. J Hematol Oncol 2017;10:159. [Crossref] [PubMed]
  20. Al-Kershi S, Bhayadia R, Ng M, et al. The stem cell-specific long noncoding RNA HOXA10-AS in the pathogenesis of KMT2A-rearranged leukemia. Blood Adv 2019;3:4252-63. [Crossref] [PubMed]
  21. Ayton PM, Cleary ML. Transformation of myeloid progenitors by MLL oncoproteins is dependent on Hoxa7 and Hoxa9. Genes Dev 2003;17:2298-307. [Crossref] [PubMed]
  22. Alexander TB, Gu Z, Iacobucci I, et al. The genetic basis and cell of origin of mixed phenotype acute leukaemia. Nature 2018;562:373-9. [Crossref] [PubMed]
doi: 10.21037/actr-24-188
Cite this article as: Thomas X. Revumenib: a new potential targeted therapy for KMT2A-rearranged acute leukemia. AME Clin Trials Rev 2025;3:14.

Download Citation