Molecular Features and Treatment Paradigms of Acute Myeloid Leukemia
Abstract
:1. Introduction
2. Somatic Mutations in AML
2.1. FLT3
2.2. NPMI Mutation
2.3. IDH
Drug | Mechanism | Setting | Major Trial |
---|---|---|---|
Midostaurin | Multi-kinase inhibitor | Newly diagnosed FLT3-mutated AML (in combination with chemotherapy) [27] | RATIFY [26] |
Gilteritinib | Highly selective, multi-kinase inhibitor | R/R FLT3-mutated AML [29] | ADMIRAL [28] |
Quizartinib | Selective kinase inhibitor | Newly diagnosed FLT3-ITD-mutated AML (in combination with chemotherapy in induction/consolidation) and as maintenance monotherapy following consolidation [35] | QuANTUM-First [34] |
Enasidenib | IDH2 inhibitor | Relapsed or Refractory AML with IDH2 mutation [107] | AG221-C-001 [85] |
Ivosidenib | IDH1 inhibitor | Newly diagnosed AML (in elderly or unable to receive intensive induction) with susceptible IDH1 mutation (alone or in combination with azacitadine), relapsed or refractory AML with susceptible IDH1 mutation [93,94,95] | AG120-C-001 [88,89], AG120-C-009 [92] |
Olutasidenib | IDH1 inhibitor | Relapsed or refractory AML with susceptible IDH1 mutation [100] | Study 2102-HEM-101 [102,103] |
2.4. TP53
2.5. KMT2A/MLL
2.6. EZH2
2.7. PHF6
2.8. U2AF1
2.9. SF3B1
2.10. SRSF2
2.11. C-KIT
2.12. KRAS/NRAS
3. Conclusions
Mutation | Clinical Trial Treatment | Patient Cohort | n | Clinical Phase | Status | Response Rate | Reference |
---|---|---|---|---|---|---|---|
TP53 | Nivolumab + decitabine + VEN | New TP53-mutated AML | 1 | I | Active, not recruiting | NCT04277442 | |
TP53 | Entrectinib + ASTX727 (decitabine + cedazuridine) | R/R AML with TP53 mutation | 12 (est) | I | Recruiting | NCT05396859 | |
TP53 | Eprenetapopt (APR-246) + AZA (maintenance) | In post- allogeneic HSCT setting, AML or MDS, with TP53 mutation | 33 | II | Completed | mOS 20.6 months (AML and MDS) | NCT03931291 [113] |
TP53 | Eprenetapopt (APR-246) + AZA | MDS, MPN, MDS/MPN, CMML, non-proliferative AML, with TP53 mutation | 52–53 | I/II | Unknown | ORR 33% in AML | NCT03588078 [112] |
TP53 | Eprenetapopt (APR-246) + AZA | MDS, MDS/MPN, CMML, oligoblastic AML, with TP53 mutation | 55 | I/II | Completed | ORR 64% and CR 36% in the AML | NCT03072043 [114] |
KMT2A, NPM1 | Revumenib (SNDX-5613) + induction/consolidation chemotherapy, and maintenance | Newly diagnosed AML with KMT2Ar, NPM1c, or NUP98r mutations | 76 (est) | I | Recruiting | NCT06226571 | |
KMT2A, NPM1 | Revumenib (SNDX-5613) + chemotherapy | R/R AML, ALL, ALAL with KMT2Ar, KMT2A amplification, NPM1c, NUP98r | 30 | I | Active, not recruiting | NCT05326516 | |
KMT2A, NPM1 | Revumenib (SNDX-5613) + “7 + 3” | New AML with NPM1 mutation or KMT2Ar | 28 (est) | I | Recruiting | NCT05886049 | |
KMT2A, NPM1 | Revumenib (SNDX-5613) + cobicistat | R/R acute leukemia (including ALL/MPAL or AML) including NUP98r, NPM1 mutation, or KMT2Ar (including pediatric population) | 413 (est) | I/II | Recruiting | NCT04065399 | |
KMT2A, NPM1 | Revumenib (SNDX-5613) + gilteritinib | R/R AML with FLT3 mutation and concurrent MLL rearrangement or NPM1 mutation, or other mutations that cause HOXA-MEIS1 overexpression | 30 (est) | I | Recruiting | NCT06222580 | |
KMT2A, NPM1 | Revumenib + venetoclax | AML with KMT2Ar, NPM1 mutation, or NUP98r (includes pediatric population) | 8 (est) | I/II | Not yet recruiting | NCT06284486 | |
KMT2A, NPM1 | Ziftomenib + AZA + VEN, ziftomenib + VEN, ziftomenib + “7 + 3” | Newly diagnosed or R/R AML with KMT2Ar or NPM1 mutation | 212 (est) | I | Recruiting | NCT05735184 | |
KMT2A, NPM1 | Ziftomenib in combination depending on cohort | R/R AML with NPM1 mutation or KMT2Ar, FLT3 mutation | 171 (est) | I | Recruiting | NCT06001788 | |
KMT2A, NPM1 | Ziftomenib | R/R AML with NPM1 mutation or KMT2Ar | 199 (est) | I/II | Recruiting | NCT04067336 | |
KMT2A, NPM1 | Ziftomenib in maintenance post-allogeneic HSCT | AML post- allogeniec HSCT with KMT2Ar or NPM1 mutation | 22 (est) | I | Not yet recruiting | NCT06440135 | |
KMT2A, NPM1 | JNJ-75276617 in combination with standard therapies depending on cohort | De novo AML, secondary AML, or R/R AML, with KMT2A or NPM1 alterations | 150 (est) | I | Recruiting | NCT05453903 | |
KMT2A, NPM1 | JNJ-75276617 with conventional chemotherapy | Pediatric and young adult patients with R/R AML with KMT2A alterations, NPM1 alterations, or nucleoporin (NUP98 or NUP214) alterations | 80 (est) | I | Not yet recruiting | NCT05521087 | |
KMT2A, NPM1 | JNJ-75276617 (Bleximenib) | R/R AML with KMT2A-r, NPM1 mutations, NUP98 alterations, or NUP214 alterations (includes pediatric population) | 350 (est) | I/II | Recruiting | NCT04811560 | |
KMT2A, NPM1 | DSP-5336 | R/R AML, ALL, or ALAL with KMT2A fusion or NPM1 mutation | 70 (est) | I/II | Recruiting | NCT04988555 | |
KMT2A, NPM1 | DS-1594b +/− chemotherapy (varying regimens) | R/R AML or ALL, with NPM1 mutation or MLLr | 17 | I/II | Completed | NCT04752163 | |
KMT2A | Pinometostat (EPZ-5676) | Advanced MLL-r leukemias | 51 | I | Completed | 1 patient with t(11;19) MLL-r AML had CR | NCT01684150, [134] |
KMT2A | Pinometostat (EPZ-5676) + azacitadine | MLL rearrangement or partial tandem duplication translocation in AML (R/R or untreated patients deferring induction) | 1 | I/II | Completed | NCT03701295 [135] | |
KIT | Avapritinib | Core binding factor AML with KIT mutation (includes pediatric population) | 50 (est) | II | Recruiting | NCT05821738 | |
KIT | Nilotinib + daunorubicin + cytarabine (“7 + 3”), then nilotinib + consolidation, then nilotinib maintenance | New AML with KIT expression and/ or mutation | 34 | II | Completed | CR/CRi 61.7% in ITT, CR/CRi 78% in evaluable patients | NCT01806571 [183] |
KIT | Imatinib (maintenance) | Newly diagnosed c-kit-positive AML patients who have received induction and post-remission treatment | 32 | II | Completed | Median PFS 52.1 months (age < 60) and median PFS 10.7 months (age ≥ 60) | NCT00509093 [178] |
KRAS/NRAS | Cobimetinib + enasidenib mesylate | R/R AML with IDH2 mutation and RAS pathway mutation | 15 (est) | I | Recruiting | NCT05441514 | |
U2AF1/SF3B1 | Emavusertib (CA-4948) | R/R AML with FLT3 mutations (previously treated with FLT3 inhibitor) or U2AF1/SF3B1 mutation, or R/R high-risk MDS with U2AF1/SF3B1 mutations | 366 (est) | I/II | Recruiting | NCT04278768 | |
SRSF2 | None | ||||||
PHF6 | None | ||||||
EZH2 | None |
Author Contributions
Funding
Conflicts of Interest
References
- Kell, J. Considerations and challenges for patients with refractory and relapsed acute myeloid leukaemia. Leuk. Res. 2016, 47, 149–160. [Google Scholar] [CrossRef] [PubMed]
- Bowman, R.L.; Busque, L.; Levine, R.L. Clonal Hematopoiesis and Evolution to Hematopoietic Malignancies. Cell Stem Cell 2018, 22, 157–170. [Google Scholar] [CrossRef] [PubMed]
- Shimony, S.; Stahl, M.; Stone, R.M. Acute myeloid leukemia: 2023 update on diagnosis, risk-stratification, and management. Am. J. Hematol. 2023, 98, 502–526. [Google Scholar] [CrossRef] [PubMed]
- Arber, D.A.; Hasserjian, R.P.; Orazi, A.; Mathews, V.; Roberts, A.W.; Schiffer, C.A.; Roug, A.S.; Cazzola, M.; Döhner, H.; Tefferi, A. Classification of myeloid neoplasms/acute leukemia: Global perspectives and the international consensus classification approach. Am. J. Hematol. 2022, 97, 514–518. [Google Scholar] [CrossRef] [PubMed]
- Döhner, H.; Weisdorf, D.J.; Bloomfield, C.D. Acute Myeloid Leukemia. N. Engl. J. Med. 2015, 373, 1136–1152. [Google Scholar] [CrossRef] [PubMed]
- Luskin, M.R.; Lee, J.-W.; Fernandez, H.F.; Abdel-Wahab, O.; Bennett, J.M.; Ketterling, R.P.; Lazarus, H.M.; Levine, R.L.; Litzow, M.R.; Paietta, E.M.; et al. Benefit of high-dose daunorubicin in AML induction extends across cytogenetic and molecular groups. Blood 2016, 127, 1551–1558. [Google Scholar] [CrossRef] [PubMed]
- Lancet, J.E.; Uy, G.L.; Cortes, J.E.; Newell, L.F.; Lin, T.L.; Ritchie, E.K.; Stuart, R.K.; Strickland, S.A.; Hogge, D.; Solomon, S.R.; 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, 2684–2692. [Google Scholar] [CrossRef] [PubMed]
- Dinardo, C.D.; Jonas, B.A.; Pullarkat, V.; Thirman, M.J.; Garcia, J.S.; Wei, A.H.; Konopleva, M.; Döhner, H.; Letai, A.; Fenaux, P.; et al. Azacitidine and Venetoclax in Previously Untreated Acute Myeloid Leukemia. N. Engl. J. Med. 2020, 383, 617–629. [Google Scholar] [CrossRef] [PubMed]
- Papaemmanuil, E.; Gerstung, M.; Bullinger, L.; Gaidzik, V.I.; Paschka, P.; Roberts, N.D.; Potter, N.E.; Heuser, M.; Thol, F.; Bolli, N.; et al. Genomic Classification and Prognosis in Acute Myeloid Leukemia. N. Engl. J. Med. 2016, 374, 2209–2221. [Google Scholar] [CrossRef]
- Garg, M.; Nagata, Y.; Kanojia, D.; Mayakonda, A.; Yoshida, K.; Keloth, S.H.; Zang, Z.J.; Okuno, Y.; Shiraishi, Y.; Chiba, K.; et al. Profiling of somatic mutations in acute myeloid leukemia with FLT3-ITD at diagnosis and relapse. Blood 2015, 126, 2491–2501. [Google Scholar] [CrossRef]
- Yoshida, K.; Sanada, M.; Shiraishi, Y.; Nowak, D.; Nagata, Y.; Yamamoto, R.; Sato, Y.; Sato-Otsubo, A.; Kon, A.; Nagasaki, M.; et al. Frequent pathway mutations of splicing machinery in myelodysplasia. Nature 2011, 478, 64–69. [Google Scholar] [CrossRef]
- Gilliland, D.G.; Griffin, J.D. The roles of FLT3 in hematopoiesis and leukemia. Blood 2002, 100, 1532–1542. [Google Scholar] [CrossRef]
- Piacibello, W.; Fubini, L.; Sanavio, F.; Brizzi, M.F.; Severino, A.; Garetto, L.; Stacchini, A.; Pegoraro, L.; Aglietta, M. Effects of human FLT3 ligand on myeloid leukemia cell growth: Heterogeneity in response and synergy with other hematopoietic growth factors. Blood 1995, 86, 4105–4114. [Google Scholar] [CrossRef] [PubMed]
- Pratz, K.W.; Levis, M. How I treat FLT3-mutated AML. Blood 2017, 129, 565–571. [Google Scholar] [CrossRef]
- Kiyoi, H.; Kawashima, N.; Ishikawa, Y. FLT3 mutations in acute myeloid leukemia: Therapeutic paradigm beyond inhibitor development. Cancer Sci. 2020, 111, 312–322. [Google Scholar] [CrossRef] [PubMed]
- Zhao, J.C.; Agarwal, S.; Ahmad, H.; Amin, K.; Bewersdorf, J.P.; Zeidan, A.M. A review of FLT3 inhibitors in acute myeloid leukemia. Blood Rev. 2022, 52, 100905. [Google Scholar] [CrossRef]
- Kiyoi, H.; Naoe, T.; Nakano, Y.; Yokota, S.; Minami, S.; Miyawaki, S.; Asou, N.; Kuriyama, K.; Jinnai, I.; Shimazaki, C.; et al. Prognostic implication of FLT3 and N-RAS gene mutations in acute myeloid leukemia. Blood 1999, 93, 3074–3080. [Google Scholar] [PubMed]
- Bacher, U.; Haferlach, C.; Kern, W.; Haferlach, T.; Schnittger, S. Prognostic relevance of FLT3-TKD mutations in AML: The combination matters—An analysis of 3082 patients. Blood 2008, 111, 2527–2537. [Google Scholar] [CrossRef]
- Yanada, M.; Matsuo, K.; Suzuki, T.; Kiyoi, H.; Naoe, T. Prognostic significance of FLT3 internal tandem duplication and tyrosine kinase domain mutations for acute myeloid leukemia: A meta-analysis. Leukemia 2005, 19, 1345–1349. [Google Scholar] [CrossRef] [PubMed]
- Li, S.; Li, N.; Chen, Y.; Zheng, Z.; Guo, Y. FLT3-TKD in the prognosis of patients with acute myeloid leukemia: A meta-analysis. Front. Oncol. 2023, 13, 1086846. [Google Scholar] [CrossRef]
- Döhner, H.; Wei, A.H.; Appelbaum, F.R.; Craddock, C.; DiNardo, C.D.; Dombret, H.; Ebert, B.L.; Fenaux, P.; Godley, L.A.; Hasserjian, R.P.; et al. Diagnosis and management of AML in adults: 2022 recommendations from an international expert panel on behalf of the ELN. Blood 2022, 140, 1345–1377. [Google Scholar] [CrossRef] [PubMed]
- Döhner, H.; Estey, E.; Grimwade, D.; Amadori, S.; Appelbaum, F.R.; Büchner, T.; Dombret, H.; Ebert, B.L.; Fenaux, P.; Larson, R.A.; et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood 2017, 129, 424–447. [Google Scholar] [CrossRef] [PubMed]
- Borthakur, G.; Kantarjian, H.; Ravandi, F.; Zhang, W.; Konopleva, M.; Wright, J.J.; Faderl, S.; Verstovsek, S.; Mathews, S.; Andreeff, M.; et al. Phase I study of sorafenib in patients with refractory or relapsed acute leukemias. Haematologica 2011, 96, 62–68. [Google Scholar] [CrossRef] [PubMed]
- Röllig, C.; Serve, H.; Hüttmann, A.; Noppeney, R.; Müller-Tidow, C.; Krug, U.; Baldus, C.D.; Brandts, C.H.; Kunzmann, V.; Einsele, H.; et al. Addition of sorafenib versus placebo to standard therapy in patients aged 60 years or younger with newly diagnosed acute myeloid leukaemia (SORAML): A multicentre, phase 2, randomised controlled trial. Lancet Oncol. 2015, 16, 1691–1699. [Google Scholar] [CrossRef] [PubMed]
- Loo, S.; Roberts, A.W.; Anstee, N.S.; Kennedy, G.A.; He, S.; Schwarer, A.P.; Enjeti, A.K.; D’Rozario, J.; Marlton, P.; Bilmon, I.A.; et al. Sorafenib plus intensive chemotherapy in newly diagnosed FLT3-ITD AML: A randomized, placebo-controlled study by the ALLG. Blood 2023, 142, 1960–1971. [Google Scholar] [CrossRef] [PubMed]
- Stone, R.M.; Mandrekar, S.J.; Sanford, B.L.; Laumann, K.; Geyer, S.; Bloomfield, C.D.; Thiede, C.; Prior, T.W.; Döhner, K.; Marcucci, G.; et al. Midostaurin plus Chemotherapy for Acute Myeloid Leukemia with a FLT3 Mutation. N. Engl. J. Med. 2017, 377, 454–464. [Google Scholar] [CrossRef]
- FDA Approves New Combination Treatment for Acute Myeloid Leukemia. Available online: https://www.fda.gov/news-events/press-announcements/fda-approves-new-combination-treatment-acute-myeloid-leukemia (accessed on 4 May 2024).
- Perl, A.E.; Martinelli, G.; Cortes, J.E.; Neubauer, A.; Berman, E.; Paolini, S.; Montesinos, P.; Baer, M.R.; Larson, R.A.; Ustun, C.; et al. Gilteritinib or Chemotherapy for Relapsed or Refractory FLT3-Mutated AML. N. Engl. J. Med. 2019, 381, 1728–1740. [Google Scholar] [CrossRef] [PubMed]
- FDA Approves Gilteritinib for Relapsed or Refractory Acute Myeloid Leukemia (AML) with a FLT3 Mutation. Available online: https://www.fda.gov/drugs/fda-approves-gilteritinib-relapsed-or-refractory-acute-myeloid-leukemia-aml-flt3-mutation (accessed on 4 May 2024).
- Pratz, K.W.; Cherry, M.; Altman, J.K.; Cooper, B.W.; Podoltsev, N.A.; Cruz, J.C.; Lin, T.L.; Schiller, G.J.; Jurcic, J.G.; Asch, A.; et al. Gilteritinib in Combination with Induction and Consolidation Chemotherapy and as Maintenance Therapy: A Phase IB Study in Patients with Newly Diagnosed AML. J. Clin. Oncol. 2023, 41, 4236–4246. [Google Scholar] [CrossRef]
- Wang, E.S.; Montesinos, P.; Minden, M.D.; Lee, J.-H.; Heuser, M.; Naoe, T.; Chou, W.-C.; Laribi, K.; Esteve, J.; Altman, J.K.; et al. Phase 3 trial of gilteritinib plus azacitidine vs. azacitidine for newly diagnosed FLT3mut+ AML ineligible for intensive chemotherapy. Blood 2022, 140, 1845–1857. [Google Scholar] [CrossRef]
- Short, N.J.; Daver, N.; Dinardo, C.D.; Kadia, T.; Nasr, L.F.; Macaron, W.; Yilmaz, M.; Borthakur, G.; Montalban-Bravo, G.; Garcia-Manero, G.; et al. Azacitidine, Venetoclax, and Gilteritinib in Newly Diagnosed and Relapsed or Refractory FLT3-Mutated AML. J. Clin. Oncol. 2024, 42, 1499–1508. [Google Scholar] [CrossRef]
- Cortes, J.E.; Khaled, S.; Martinelli, G.; Perl, A.E.; Ganguly, S.; Russell, N.; Krämer, A.; Dombret, H.; Hogge, D.; Jonas, B.A.; et al. Quizartinib versus salvage chemotherapy in relapsed or refractory FLT3-ITD acute myeloid leukaemia (QuANTUM-R): A multicentre, randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2019, 20, 984–997. [Google Scholar] [CrossRef]
- Erba, H.P.; Montesinos, P.; Kim, H.J.; Patkowska, E.; Vrhovac, R.; Žák, P.; Wang, P.-N.; Mitov, T.; Hanyok, J.; Kamel, Y.M.; et al. Quizartinib plus chemotherapy in newly diagnosed patients with FLT3-internal-tandem-duplication-positive acute myeloid leukaemia (QuANTUM-First): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2023, 401, 1571–1583. [Google Scholar] [CrossRef]
- FDA Approves Quizartinib for Newly Diagnosed Acute Myeloid Leukemia. Available online: https://www.fda.gov/drugs/drug-approvals-and-databases/fda-approves-quizartinib-newly-diagnosed-acute-myeloid-leukemia (accessed on 4 May 2024).
- Randhawa, J.K.; Kantarjian, H.M.; Borthakur, G.; Thompson, P.A.; Konopleva, M.; Daver, N.; Pemmaraju, N.; Jabbour, E.; Kadia, T.M.; Estrov, Z.; et al. Results of a Phase II Study of Crenolanib in Relapsed/Refractory Acute Myeloid Leukemia Patients (Pts) with Activating FLT3 Mutations. Blood 2014, 124, 389. [Google Scholar] [CrossRef]
- Cortes, J.E.; Kantarjian, H.M.; Kadia, T.M.; Borthakur, G.; Konopleva, M.; Garcia-Manero, G.; Daver, N.G.; Pemmaraju, N.; Jabbour, E.; Estrov, Z.; et al. Crenolanib besylate, a type I pan-FLT3 inhibitor, to demonstrate clinical activity in multiply relapsed FLT3-ITD and D835 AML. J. Clin. Oncol. 2016, 34 (Suppl. S15), 7008. [Google Scholar] [CrossRef]
- Chen, Y.-B.; Li, S.; Lane, A.A.; Connolly, C.; Del Rio, C.; Valles, B.; Curtis, M.; Ballen, K.; Cutler, C.; Dey, B.R.; et al. Phase I trial of maintenance sorafenib after allogeneic hematopoietic stem cell transplantation for fms-like tyrosine kinase 3 internal tandem duplication acute myeloid leukemia. Biol. Blood Marrow Transplant. 2014, 20, 2042–2048. [Google Scholar] [CrossRef]
- Burchert, A.; Bug, G.; Fritz, L.V.; Finke, J.; Stelljes, M.; Röllig, C.; Wollmer, E.; Wäsch, R.; Bornhäuser, M.; Berg, T.; et al. Sorafenib Maintenance After Allogeneic Hematopoietic Stem Cell Transplantation for Acute Myeloid Leukemia with FLT3-Internal Tandem Duplication Mutation (SORMAIN). J. Clin. Oncol. 2020, 38, 2993–3002. [Google Scholar] [CrossRef]
- Xuan, L.; Wang, Y.; Huang, F.; Fan, Z.; Xu, Y.; Sun, J.; Xu, N.; Deng, L.; Li, X.; Liang, X.; et al. Sorafenib maintenance in patients with FLT3-ITD acute myeloid leukaemia undergoing allogeneic haematopoietic stem-cell transplantation: An open-label, multicentre, randomised phase 3 trial. Lancet Oncol. 2020, 21, 1201–1212. [Google Scholar] [CrossRef]
- Maziarz, R.T.; Levis, M.; Patnaik, M.M.; Scott, B.L.; Mohan, S.R.; Deol, A.; Rowley, S.D.; Kim, D.D.H.; Hernandez, D.; Rajkhowa, T.; et al. Midostaurin after allogeneic stem cell transplant in patients with FLT3-internal tandem duplication-positive acute myeloid leukemia. Bone Marrow Transplant. 2021, 56, 1180–1189. [Google Scholar] [CrossRef]
- Levis, M.J.; Hamadani, M.; Logan, B.R.; Rosales, M.; Delgado, D.; Bahceci, E.; Devine, S.; Horowitz, M.M.; Chen, Y.-B. BMT CTN Protocol 1506: A Phase 3 Trial of Gilteritinib As Maintenance Therapy after Allogeneic Hematopoietic Stem Cell Transplantation in Patients with FLT3-ITD+ AML. Blood 2019, 134 (Suppl. S1), 4602. [Google Scholar] [CrossRef]
- Levis, M.J.; Hamadani, M.; Logan, B.; Jones, R.J.; Singh, A.K.; Litzow, M.; Wingard, J.R.; Papadopoulos, E.B.; Perl, A.E.; Soiffer, R.J.; et al. Gilteritinib as Post-Transplant Maintenance for AML with Internal Tandem Duplication Mutation of FLT3. J. Clin. Oncol. 2024, 42, 1766–1775. [Google Scholar] [CrossRef]
- Grob, T.; Sanders, M.A.; Vonk, C.M.; Kavelaars, F.G.; Rijken, M.; Hanekamp, D.W.; Gradowska, P.L.; Cloos, J.; Fløisand, Y.; Kooy, M.v.M.; et al. Prognostic Value of FLT3-Internal Tandem Duplication Residual Disease in Acute Myeloid Leukemia. J. Clin. Oncol. 2023, 41, 756–765. [Google Scholar] [CrossRef] [PubMed]
- Dillon, L.W.; Gui, G.; Page, K.M.; Ravindra, N.; Wong, Z.C.; Andrew, G.; Mukherjee, D.; Zeger, S.L.; El Chaer, F.; Spellman, S.; et al. DNA Sequencing to Detect Residual Disease in Adults with Acute Myeloid Leukemia Prior to Hematopoietic Cell Transplant. JAMA 2023, 329, 745–755. [Google Scholar] [CrossRef] [PubMed]
- Sandmaier, B.M.; Khaled, S.; Oran, B.; Gammon, G.; Trone, D.; Frankfurt, O. Results of a phase 1 study of quizartinib as maintenance therapy in subjects with acute myeloid leukemia in remission following allogeneic hematopoietic stem cell transplant. Am. J. Hematol. 2018, 93, 222–231. [Google Scholar] [CrossRef] [PubMed]
- Ruglioni, M.; Crucitta, S.; Luculli, G.I.; Tancredi, G.; Del Giudice, M.L.; Mechell, S.; Galimberti, S.; Danesi, R.; Del Re, M. Understanding mechanisms of resistance to FLT3 inhibitors in adult FLT3-mutated Acute Myeloid Leukemia (AML) to guide treatment strategy. Crit. Rev. Oncol. Hematol. 2024, 201, 104424. [Google Scholar] [CrossRef] [PubMed]
- Falini, B.; Brunetti, L.; Sportoletti, P.; Martelli, M.P. NPM1-mutated acute myeloid leukemia: From bench to bedside. Blood 2020, 136, 1707–1721. [Google Scholar] [CrossRef] [PubMed]
- Brunetti, L.; Gundry, M.C.; Sorcini, D.; Guzman, A.G.; Huang, Y.-H.; Ramabadran, R.; Gionfriddo, I.; Mezzasoma, F.; Milano, F.; Nabet, B.; et al. Mutant NPM1 Maintains the Leukemic State through HOX Expression. Cancer Cell 2018, 34, 499–512.e9. [Google Scholar] [CrossRef] [PubMed]
- Grisendi, S.; Mecucci, C.; Falini, B.; Pandolfi, P.P. Nucleophosmin and cancer. Nat. Rev. Cancer 2006, 6, 493–505. [Google Scholar] [CrossRef]
- Pianigiani, G.; Gagliardi, A.; Mezzasoma, F.; Rocchio, F.; Tini, V.; Bigerna, B.; Sportoletti, P.; Caruso, S.; Marra, A.; Peruzzi, S.; et al. Prolonged XPO1 inhibition is essential for optimal antileukemic activity in NPM1-mutated AML. Blood Adv. 2022, 6, 5938–5949. [Google Scholar] [CrossRef] [PubMed]
- Khoury, J.D.; Solary, E.; Abla, O.; Akkari, Y.; Alaggio, R.; Apperley, J.F.; Bejar, R.; Berti, E.; Busque, L.; Chan, J.K.C.; et al. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Myeloid and Histiocytic/Dendritic Neoplasms. Leukemia 2022, 36, 1703–1719. [Google Scholar] [CrossRef]
- Ranieri, R.; Pianigiani, G.; Sciabolacci, S.; Perriello, V.M.; Marra, A.; Cardinali, V.; Pierangeli, S.; Milano, F.; Gionfriddo, I.; Brunetti, L.; et al. Current status and future perspectives in targeted therapy of NPM1-mutated AML. Leukemia 2022, 36, 2351–2367. [Google Scholar] [CrossRef]
- Wang, R.; Xu, P.; Chang, L.-L.; Zhang, S.-Z.; Zhu, H.-H. Targeted therapy in NPM1-mutated AML: Knowns and unknowns. Front. Oncol. 2022, 12, 972606. [Google Scholar] [CrossRef] [PubMed]
- Hing, Z.A.; Fung, H.Y.J.; Ranganathan, P.; Mitchell, S.; El-Gamal, D.; Woyach, J.A.; Williams, K.; Goettl, V.M.; Smith, J.; Yu, X.; et al. Next-generation XPO1 inhibitor shows improved efficacy and in vivo tolerability in hematological malignancies. Leukemia 2016, 30, 2364–2372. [Google Scholar] [CrossRef]
- Garzon, R.; Savona, M.; Baz, R.; Andreeff, M.; Gabrail, N.; Gutierrez, M.; Savoie, L.; Mau-Sorensen, P.M.; Wagner-Johnston, N.; Yee, K.; et al. A phase 1 clinical trial of single-agent selinexor in acute myeloid leukemia. Blood 2017, 129, 3165–3174. [Google Scholar] [CrossRef]
- Bhatnagar, B.; Zhao, Q.; Mims, A.S.; Vasu, S.; Behbehani, G.K.; Larkin, K.; Blachly, J.S.; Blum, W.; Klisovic, R.B.; Ruppert, A.S.; et al. Selinexor in combination with decitabine in patients with acute myeloid leukemia: Results from a phase 1 study. Leuk. Lymphoma 2020, 61, 387–396. [Google Scholar] [CrossRef]
- Wang, A.Y.; Weiner, H.; Green, M.; Chang, H.; Fulton, N.; Larson, R.A.; Odenike, O.; Artz, A.S.; Bishop, M.R.; Godley, L.A.; et al. A phase I study of selinexor in combination with high-dose cytarabine and mitoxantrone for remission induction in patients with acute myeloid leukemia. J. Hematol. Oncol. 2018, 11, 4. [Google Scholar] [CrossRef]
- Fiedler, W.; Chromik, J.; Amberg, S.; Kebenko, M.; Thol, F.; Schlipfenbacher, V.; Wilke, A.C.; Modemann, F.; Janning, M.; Serve, H.; et al. A Phase II study of selinexor plus cytarabine and idarubicin in patients with relapsed/refractory acute myeloid leukaemia. Br. J. Haematol. 2020, 190, e169–e173. [Google Scholar] [CrossRef] [PubMed]
- Sweet, K.; Bhatnagar, B.; Döhner, H.; Donnellan, W.; Frankfurt, O.; Heuser, M.; Kota, V.; Liu, H.; Raffoux, E.; Roboz, G.J.; et al. A 2:1 randomized, open-label, phase II study of selinexor vs. physician’s choice in older patients with relapsed or refractory acute myeloid leukemia. Leuk. Lymphoma 2021, 62, 3192–3203. [Google Scholar] [CrossRef]
- Etchin, J.; Berezovskaya, A.; Conway, A.S.; Galinsky, I.A.; Stone, R.M.; Baloglu, E.; Senapedis, W.; Landesman, Y.; Kauffman, M.; Shacham, S.; et al. KPT-8602, a second-generation inhibitor of XPO1-mediated nuclear export, is well tolerated and highly active against AML blasts and leukemia-initiating cells. Leukemia 2017, 31, 143–150. [Google Scholar] [CrossRef]
- Wei, A.H.; Montesinos, P.; Ivanov, V.; DiNardo, C.D.; Novak, J.; Laribi, K.; Kim, I.; Stevens, D.A.; Fiedler, W.; Pagoni, M.; et al. Venetoclax plus LDAC for newly diagnosed AML ineligible for intensive chemotherapy: A phase 3 randomized placebo-controlled trial. Blood 2020, 135, 2137–2145. [Google Scholar] [CrossRef] [PubMed]
- DiNardo, C.D.; Lachowiez, C.A.; Takahashi, K.; Loghavi, S.; Xiao, L.; Kadia, T.; Daver, N.; Adeoti, M.; Short, N.J.; Sasaki, K.; et al. Venetoclax Combined with FLAG-IDA Induction and Consolidation in Newly Diagnosed and Relapsed or Refractory Acute Myeloid Leukemia. J. Clin. Oncol. 2021, 39, 2768–2778. [Google Scholar] [CrossRef]
- Chua, C.C.; Roberts, A.W.; Reynolds, J.; Fong, C.Y.; Ting, S.B.; Salmon, J.M.; MacRaild, S.; Ivey, A.; Tiong, I.S.; Fleming, S.; et al. Chemotherapy and Venetoclax in Elderly Acute Myeloid Leukemia Trial (CAVEAT): A Phase Ib Dose-Escalation Study of Venetoclax Combined with Modified Intensive Chemotherapy. J. Clin. Oncol. 2020, 38, 3506–3517. [Google Scholar] [CrossRef] [PubMed]
- Lachowiez, C.A.; Loghavi, S.; Kadia, T.M.; Daver, N.; Borthakur, G.; Pemmaraju, N.; Naqvi, K.; Alvarado, Y.; Yilmaz, M.; Short, N.; et al. Outcomes of older patients with NPM1-mutated AML: Current treatments and the promise of venetoclax-based regimens. Blood Adv. 2020, 4, 1311–1320. [Google Scholar] [CrossRef] [PubMed]
- Kühn, M.W.M.; Song, E.; Feng, Z.; Sinha, A.; Chen, C.-W.; Deshpande, A.J.; Cusan, M.; Farnoud, N.; Mupo, A.; Grove, C.; et al. Targeting Chromatin Regulators Inhibits Leukemogenic Gene Expression in NPM1 Mutant Leukemia. Cancer Discov. 2016, 6, 1166–1181. [Google Scholar] [CrossRef] [PubMed]
- van der Lee, D.I.; Reijmers, R.M.; Honders, M.W.; Hagedoorn, R.S.; de Jong, R.C.; Kester, M.G.; van der Steen, D.M.; de Ru, A.H.; Kweekel, C.; Bijen, H.M.; et al. Mutated nucleophosmin 1 as immunotherapy target in acute myeloid leukemia. J. Clin. Investig. 2019, 129, 774–785. [Google Scholar] [CrossRef] [PubMed]
- Xie, G.; Ivica, N.A.; Jia, B.; Li, Y.; Dong, H.; Liang, Y.; Brown, D.; Romee, R.; Chen, J. CAR-T cells targeting a nucleophosmin neoepitope exhibit potent specific activity in mouse models of acute myeloid leukaemia. Nat. Biomed. Eng. 2021, 5, 399–413. [Google Scholar] [CrossRef] [PubMed]
- Falini, B.; Dillon, R. Criteria for Diagnosis and Molecular Monitoring of NPM1-Mutated AML. Blood Cancer Discov. 2024, 5, 8–20. [Google Scholar] [CrossRef] [PubMed]
- Krönke, J.; Schlenk, R.F.; Jensen, K.-O.; Tschürtz, F.; Corbacioglu, A.; Gaidzik, V.I.; Paschka, P.; Onken, S.; Eiwen, K.; Habdank, M.; et al. Monitoring of minimal residual disease in NPM1-mutated acute myeloid leukemia: A study from the German-Austrian acute myeloid leukemia study group. J. Clin. Oncol. 2011, 29, 2709–2716. [Google Scholar] [CrossRef] [PubMed]
- Orvain, C.; Bertoli, S.; Peterlin, P.; Desbrosses, Y.; Dumas, P.-Y.; Iat, A.; Marie Ann Hospital; Carre, M.; Tavernier, E.; Bouvier, A.; et al. Molecular Relapse after First-Line Intensive Therapy in Patients with Core-Binding Factor and NPM1-Mutated Acute Myeloid Leukemia—A Filo Study. Blood 2023, 142 (Suppl. S1), 421. [Google Scholar] [CrossRef]
- Othman, J.; Potter, N.; Ivey, A.; Jovanovic, J.; Freeman, S.D.; Gilkes, A.; Thomas, I.; Johnson, S.; Canham, J.; Cavenagh, J.; et al. The Benefit of Allogeneic Transplant in 1st Complete Remission in NPM1 Mutated AML with or without FLT3 ITD Is Restricted to Those Testing MRD Positive after Induction—An Analysis of the UK NCRI AML17 and AML19 Studies. Blood 2023, 142 (Suppl. S1), 425. [Google Scholar] [CrossRef]
- Xue, E.; Tresoldi, C.; Sala, E.; Crippa, A.; Mazzi, B.; Greco, R.; Messina, C.; Carrabba, M.G.; Stanghellini, M.T.L.; Marktel, S.; et al. Longitudinal qPCR monitoring of nucleophosmin 1 mutations after allogeneic hematopoietic stem cell transplantation to predict AML relapse. Bone Marrow Transplant. 2016, 51, 466–469. [Google Scholar] [CrossRef]
- Heath, E.M.; Chan, S.M.; Minden, M.D.; Murphy, T.; Shlush, L.I.; Schimmer, A.D. Biological and clinical consequences of NPM1 mutations in AML. Leukemia 2017, 31, 798–807. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [PubMed]
- Mardis, E.R.; Ding, L.; Dooling, D.J.; Larson, D.E.; McLellan, M.D.; Chen, K.; Koboldt, D.C.; Fulton, R.S.; Delehaunty, K.D.; McGrath, S.D.; et al. Recurring mutations found by sequencing an acute myeloid leukemia genome. N. Engl. J. Med. 2009, 361, 1058–1066. [Google Scholar] [CrossRef] [PubMed]
- DiNardo, C.D.; Ravandi, F.; Agresta, S.; Konopleva, M.; Takahashi, K.; Kadia, T.; Routbort, M.; Patel, K.P.; Mark, B.; Pierce, S.; et al. Characteristics, clinical outcome, and prognostic significance of IDH mutations in AML. Am. J. Hematol. 2015, 90, 732–736. [Google Scholar] [CrossRef] [PubMed]
- Ward, P.S.; Patel, J.; Wise, D.R.; Abdel-Wahab, O.; Bennett, B.D.; Coller, H.A.; Cross, J.R.; Fantin, V.R.; Hedvat, C.V.; Perl, A.E.; et al. The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting α-ketoglutarate to 2-hydroxyglutarate. Cancer Cell 2010, 17, 225–234. [Google Scholar] [CrossRef] [PubMed]
- Chou, F.-J.; Liu, Y.; Lang, F.; Yang, C. D-2-Hydroxyglutarate in Glioma Biology. Cells 2021, 10, 2345. [Google Scholar] [CrossRef] [PubMed]
- McMurry, H.; Fletcher, L.; Traer, E. IDH Inhibitors in AML—Promise and Pitfalls. Curr. Hematol. Malig. Rep. 2021, 16, 207–217. [Google Scholar] [CrossRef] [PubMed]
- Ragon, B.K.; DiNardo, C.D. Targeting IDH1 and IDH2 Mutations in Acute Myeloid Leukemia. Curr. Hematol. Malig. Rep. 2017, 12, 537–546. [Google Scholar] [CrossRef] [PubMed]
- Figueroa, M.E.; Abdel-Wahab, O.; Lu, C.; Ward, P.S.; Patel, J.; Shih, A.; Li, Y.; Bhagwat, N.; VasanthaKumar, A.; Fernandez, H.F.; et al. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell 2010, 18, 553–567. [Google Scholar] [CrossRef] [PubMed]
- Yen, K.; Travins, J.; Wang, F.; David, M.D.; Artin, E.; Straley, K.; Padyana, A.; Gross, S.; DeLaBarre, B.; Tobin, E.; et al. AG-221, a First-in-Class Therapy Targeting Acute Myeloid Leukemia Harboring Oncogenic IDH2 Mutations. Cancer Discov. 2017, 7, 478–493. [Google Scholar] [CrossRef]
- Wang, F.; Travins, J.; DeLaBarre, B.; Penard-Lacronique, V.; Schalm, S.; Hansen, E.; Straley, K.; Kernytsky, A.; Liu, W.; Gliser, C.; et al. Targeted inhibition of mutant IDH2 in leukemia cells induces cellular differentiation. Science 2013, 340, 622–626. [Google Scholar] [CrossRef]
- Stein, E.M.; Dinardo, C.D.; Pollyea, D.A.; Fathi, A.T.; Roboz, G.J.; Altman, J.K.; Stone, R.M.; DeAngelo, D.J.; Levine, R.L.; Flinn, I.W.; et al. Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia. Blood 2017, 130, 722–731. [Google Scholar] [CrossRef]
- Pollyea, D.A.; Tallman, M.S.; de Botton, S.; Kantarjian, H.M.; Collins, R.; Stein, A.S.; Frattini, M.G.; Xu, Q.; Tosolini, A.; See, W.L.; et al. Enasidenib, an inhibitor of mutant IDH2 proteins, induces durable remissions in older patients with newly diagnosed acute myeloid leukemia. Leukemia 2019, 33, 2575–2584. [Google Scholar] [CrossRef]
- DiNardo, C.D.; Schuh, A.C.; Stein, E.M.; Montesinos, P.; Wei, A.H.; de Botton, S.; Zeidan, A.M.; Fathi, A.T.; Kantarjian, H.M.; Bennett, J.M.; et al. Enasidenib plus azacitidine versus azacitidine alone in patients with newly diagnosed, mutant-IDH2 acute myeloid leukaemia (AG221-AML-005): A single-arm, phase 1b and randomised, phase 2 trial. Lancet Oncol. 2021, 22, 1597–1608. [Google Scholar] [CrossRef]
- DiNardo, C.D.; Stein, E.M.; de Botton, S.; Roboz, G.J.; Altman, J.K.; Mims, A.S.; Swords, R.; Collins, R.H.; Mannis, G.N.; Pollyea, D.A.; et al. Durable Remissions with Ivosidenib in IDH1-Mutated Relapsed or Refractory AML. N. Engl. J. Med. 2018, 378, 2386–2398. [Google Scholar] [CrossRef]
- Roboz, G.J.; DiNardo, C.D.; Stein, E.M.; de Botton, S.; Mims, A.S.; Prince, G.T.; Altman, J.K.; Arellano, M.L.; Donnellan, W.; Erba, H.P.; et al. Ivosidenib induces deep durable remissions in patients with newly diagnosed IDH1-mutant acute myeloid leukemia. Blood 2020, 135, 463–471. [Google Scholar] [CrossRef]
- Stein, E.M.; DiNardo, C.D.; Fathi, A.T.; Mims, A.S.; Pratz, K.W.; Savona, M.R.; Stein, A.S.; Stone, R.M.; Winer, E.S.; Seet, C.S.; et al. Ivosidenib or enasidenib combined with intensive chemotherapy in patients with newly diagnosed AML: A phase 1 study. Blood 2021, 137, 1792–1803. [Google Scholar] [CrossRef]
- DiNardo, C.D.; Stein, A.S.; Stein, E.M.; Fathi, A.T.; Frankfurt, O.; Schuh, A.C.; Döhner, H.; Martinelli, G.; Patel, P.A.; Raffoux, E.; et al. Mutant Isocitrate Dehydrogenase 1 Inhibitor Ivosidenib in Combination with Azacitidine for Newly Diagnosed Acute Myeloid Leukemia. J. Clin. Oncol. 2021, 39, 57–65. [Google Scholar] [CrossRef]
- Montesinos, P.; Recher, C.; Vives, S.; Zarzycka, E.; Wang, J.; Bertani, G.; Heuser, M.; Calado, R.T.; Schuh, A.C.; Yeh, S.-P.; et al. Ivosidenib and Azacitidine in IDH1-Mutated Acute Myeloid Leukemia. N. Engl. J. Med. 2022, 386, 1519–1531. [Google Scholar] [CrossRef]
- FDA Approves Ivosidenib in Combination with Azacitidine for Newly Diagnosed Acute Myeloid Leukemia. Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-ivosidenib-combination-azacitidine-newly-diagnosed-acute-myeloid-leukemia (accessed on 4 May 2024).
- FDA Approves Ivosidenib for Relapsed or Refractory Acute Myeloid Leukemia. Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-ivosidenib-relapsed-or-refractory-acute-myeloid-leukemia (accessed on 4 May 2024).
- FDA Approves Ivosidenib as First-Line Treatment for AML with IDH1 Mutation. Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-ivosidenib-first-line-treatment-aml-idh1-mutation (accessed on 4 May 2024).
- de Botton, S.; Montesinos, P.; Schuh, A.C.; Papayannidis, C.; Vyas, P.; Wei, A.H.; Ommen, H.; Semochkin, S.; Kim, H.-J.; Larson, R.A.; et al. Enasidenib vs. conventional care in older patients with late-stage mutant-IDH2 relapsed/refractory AML: A randomized phase 3 trial. Blood 2023, 141, 156–167. [Google Scholar] [CrossRef]
- IDHIFA (Enasidenib Mesylate)—Market Withdrawal and Continued Access. Available online: https://recalls-rappels.canada.ca/en/alert-recall/idhifa-enasidenib-mesylate-market-withdrawal-and-continued-access (accessed on 4 May 2024).
- Fruchtman, H.; Avigan, Z.M.; Waksal, J.A.; Brennan, N.; Mascarenhas, J.O. Management of isocitrate dehydrogenase 1/2 mutated acute myeloid leukemia. Leukemia 2024, 38, 927–935. [Google Scholar] [CrossRef] [PubMed]
- Venugopal, S.; Watts, J. Olutasidenib: From bench to bedside. Blood Adv. 2023, 7, 4358–4365. [Google Scholar] [CrossRef]
- FDA Approves Olutasidenib for Relapsed or Refractory Acute Myeloid Leukemia with a Susceptible IDH1 Mutation. Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-olutasidenib-relapsed-or-refractory-acute-myeloid-leukemia-susceptible-idh1-mutation (accessed on 4 May 2024).
- Watts, J.M.; Baer, M.R.; Yang, J.; Prebet, T.; Lee, S.; Schiller, G.J.; Dinner, S.; Pigneux, A.; Montesinos, P.; Wang, E.S.; et al. Olutasidenib (FT-2102), an IDH1m Inhibitor As a Single Agent or in Combination with Azacitidine, Induces Deep Clinical Responses with Mutation Clearance in Patients with Acute Myeloid Leukemia Treated in a Phase 1 Dose Escalation and Expansion Study. Blood 2019, 134 (Suppl. S1), 231. [Google Scholar] [CrossRef]
- Watts, J.M.; Baer, M.R.; Yang, J.; Prebet, T.; Lee, S.; Schiller, G.J.; Dinner, S.N.; Pigneux, A.; Montesinos, P.; Wang, E.S.; et al. Olutasidenib alone or with azacitidine in IDH1-mutated acute myeloid leukaemia and myelodysplastic syndrome: Phase 1 results of a phase 1/2 trial. Lancet Haematol. 2023, 10, e46–e58. [Google Scholar] [CrossRef] [PubMed]
- de Botton, S.; Fenaux, P.; Yee, K.W.; Récher, C.; Wei, A.H.; Montesinos, P.; Taussig, D.C.; Pigneux, A.; Braun, T.; Curti, A.; et al. Olutasidenib (FT-2102) induces durable complete remissions in patients with relapsed or refractory IDH1-mutated AML. Blood Adv. 2023, 7, 3117–3127. [Google Scholar] [CrossRef]
- Fathi, A.T.; Kim, H.T.; Soiffer, R.J.; Levis, M.J.; Li, S.; Kim, A.S.; DeFilipp, Z.; El-Jawahri, A.; McAfee, S.L.; Brunner, A.M.; et al. Multicenter Phase I Trial of Ivosidenib as Maintenance Treatment Following Allogeneic Hematopoietic Cell Transplantation for IDH1-Mutated Acute Myeloid Leukemia. Clin. Cancer Res. 2023, 29, 2034–2042. [Google Scholar] [CrossRef]
- Fathi, A.T.; Kim, H.T.; Soiffer, R.J.; Levis, M.J.; Li, S.; Kim, A.S.; Mims, A.S.; DeFilipp, Z.; El-Jawahri, A.; McAfee, S.L.; et al. Enasidenib as maintenance following allogeneic hematopoietic cell transplantation for IDH2-mutated myeloid malignancies. Blood Adv. 2022, 6, 5857–5865. [Google Scholar] [CrossRef]
- Issa, G.C.; DiNardo, C.D. Acute myeloid leukemia with IDH1 and IDH2 mutations: 2021 treatment algorithm. Blood Cancer J. 2021, 11, 107. [Google Scholar] [CrossRef]
- FDA Granted Regular Approval to Enasidenib for the Treatment of Relapsed or Refractory AML. Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-granted-regular-approval-enasidenib-treatment-relapsed-or-refractory-aml (accessed on 4 May 2024).
- Yan, B.; Claxton, D.; Huang, S.; Qiu, Y. AML chemoresistance: The role of mutant TP53 subclonal expansion and therapy strategy. Exp. Hematol. 2020, 87, 13–19. [Google Scholar] [CrossRef]
- Daver, N.G.; Maiti, A.; Kadia, T.M.; Vyas, P.; Majeti, R.; Wei, A.H.; Garcia-Manero, G.; Craddock, C.; Sallman, D.A.; Kantarjian, H.M. TP53-Mutated Myelodysplastic Syndrome and Acute Myeloid Leukemia: Biology, Current Therapy, and Future Directions. Cancer Discov. 2022, 12, 2516–2529. [Google Scholar] [CrossRef]
- Kim, K.; Maiti, A.; Loghavi, S.; Pourebrahim, R.; Kadia, T.M.; Rausch, C.R.; Furudate, K.; Daver, N.G.; Alvarado, Y.; Ohanian, M.; et al. Outcomes of TP53-mutant acute myeloid leukemia with decitabine and venetoclax. Cancer 2021, 127, 3772–3781. [Google Scholar] [CrossRef]
- Lehmann, S.; Bykov, V.J.; Ali, D.; Andrén, O.; Cherif, H.; Tidefelt, U.; Uggla, B.; Yachnin, J.; Juliusson, G.; Moshfegh, A.; 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, 3633–3639. [Google Scholar] [CrossRef] [PubMed]
- Cluzeau, T.; Sebert, M.; Rahmé, R.; Cuzzubbo, S.; Lehmann-Che, J.; Madelaine, I.; Peterlin, P.; Bève, B.; Attalah, H.; Chermat, F.; 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, 39, 1575–1583. [Google Scholar] [CrossRef] [PubMed]
- Mishra, A.; Tamari, R.; DeZern, A.E.; Byrne, M.T.; Gooptu, M.; Chen, Y.-B.; Deeg, H.J.; Sallman, D.; Gallacher, P.; Wennborg, A.; et al. Eprenetapopt Plus Azacitidine After Allogeneic Hematopoietic Stem-Cell Transplantation for TP53-Mutant Acute Myeloid Leukemia and Myelodysplastic Syndromes. J. Clin. Oncol. 2022, 40, 3985–3993. [Google Scholar] [CrossRef] [PubMed]
- Sallman, D.A.; DeZern, A.E.; Garcia-Manero, G.; Steensma, D.P.; Roboz, G.J.; Sekeres, M.A.; Cluzeau, T.; Sweet, K.L.; McLemore, A.; McGraw, K.L.; et al. Eprenetapopt (APR-246) and Azacitidine in TP53-Mutant Myelodysplastic Syndromes. J. Clin. Oncol. 2021, 39, 1584–1594. [Google Scholar] [CrossRef] [PubMed]
- Daver, N.G.; Vyas, P.; Kambhampati, S.; Al Malki, M.M.; Larson, R.A.; Asch, A.S.; Mannis, G.; Chai-Ho, W.; Tanaka, T.N.; Bradley, T.J.; et al. Tolerability and Efficacy of the Anticluster of Differentiation 47 Antibody Magrolimab Combined with Azacitidine in Patients with Previously Untreated AML: Phase Ib Results. J. Clin. Oncol. 2023, 41, 4893–4904. [Google Scholar] [CrossRef] [PubMed]
- Gilead Discontinues Phase 3 ENHANCE-2 Trial of Magrolimab Plus Azacitidine in TP53-Mutant AML. Available online: https://www.onclive.com/view/gilead-discontinues-phase-3-enhance-2-trial-of-magrolimab-plus-azacitidine-in-tp53-mutant-aml (accessed on 4 May 2024).
- Gilead Statement on Discontinuation of Phase 3 ENHANCE-3 Study in AML. Available online: https://www.gilead.com/news-and-press/company-statements/gilead-statement-on-discontinuation-of-phase-3-enhance-3-study-in-aml (accessed on 4 May 2024).
- Testa, U.; Riccioni, R.; Militi, S.; Coccia, E.; Stellacci, E.; Samoggia, P.; Latagliata, R.; Mariani, G.; Rossini, A.; Battistini, A.; et al. Elevated expression of IL-3Rα in acute myelogenous leukemia is associated with enhanced blast proliferation, increased cellularity, and poor prognosis. Blood 2002, 100, 2980–2988. [Google Scholar] [CrossRef] [PubMed]
- Al-Hussaini, M.; Rettig, M.P.; Ritchey, J.K.; Karpova, D.; Uy, G.L.; Eissenberg, L.G.; Gao, F.; Eades, W.C.; Bonvini, E.; Chichili, G.R.; et al. Targeting CD123 in acute myeloid leukemia using a T-cell-directed dual-affinity retargeting platform. Blood 2016, 127, 122–131. [Google Scholar] [CrossRef] [PubMed]
- Vadakekolathu, J.; Lai, C.; Reeder, S.; Church, S.E.; Hood, T.; Lourdusamy, A.; Rettig, M.P.; Aldoss, I.; Advani, A.S.; Godwin, J.; et al. TP53 abnormalities correlate with immune infiltration and associate with response to flotetuzumab immunotherapy in AML. Blood Adv. 2020, 4, 5011–5024. [Google Scholar] [CrossRef] [PubMed]
- Vadakekolathu, J.; Minden, M.D.; Hood, T.; Church, S.E.; Reeder, S.; Altmann, H.; Sullivan, A.H.; Viboch, E.J.; Patel, T.; Ibrahimova, N.; et al. Immune landscapes predict chemotherapy resistance and immunotherapy response in acute myeloid leukemia. Sci. Transl. Med. 2020, 12, 546. [Google Scholar] [CrossRef]
- Uy, G.L.; Aldoss, I.; Foster, M.C.; Sayre, P.H.; Wieduwilt, M.J.; Advani, A.S.; Godwin, J.E.; Arellano, M.L.; Sweet, K.L.; Emadi, A.; et al. Flotetuzumab as salvage immunotherapy for refractory acute myeloid leukemia. Blood 2021, 137, 751–762. [Google Scholar] [CrossRef] [PubMed]
- Muntean, A.G.; Hess, J.L. The pathogenesis of mixed-lineage leukemia. Annu. Rev. Pathol. 2012, 7, 283–301. [Google Scholar] [CrossRef]
- Thiel, A.T.; Blessington, P.; Zou, T.; Feather, D.; Wu, X.; Yan, J.; Zhang, H.; Liu, Z.; Ernst, P.; Koretzky, G.A.; et al. MLL-AF9-induced leukemogenesis requires coexpression of the wild-type Mll allele. Cancer Cell 2010, 17, 148–159. [Google Scholar] [CrossRef] [PubMed]
- Krivtsov, A.V.; Armstrong, S.A. MLL translocations, histone modifications and leukaemia stem-cell development. Nat. Rev. Cancer 2007, 7, 823–833. [Google Scholar] [CrossRef] [PubMed]
- Issa, G.C.; Aldoss, I.; DiPersio, J.; Cuglievan, B.; Stone, R.; Arellano, M.; Thirman, M.J.; Patel, M.R.; Dickens, D.S.; Shenoy, S.; et al. The menin inhibitor revumenib in KMT2A-rearranged or NPM1-mutant leukaemia. Nature 2023, 615, 920–924. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.; Kantarjian, H.; Pierce, S.; Faderl, S.; O’Brien, S.; Qiao, W.; Abruzzo, L.; de Lima, M.; Kebriaei, P.; Jabbour, E.; et al. Prognostic significance of 11q23 aberrations in adult acute myeloid leukemia and the role of allogeneic stem cell transplantation. Leukemia 2013, 27, 836–842. [Google Scholar] [CrossRef] [PubMed]
- Bill, M.; Mrózek, K.; Kohlschmidt, J.; Eisfeld, A.K.; Walker, C.J.; Nicolet, D.; Papaioannou, D.; Blachly, J.S.; Orwick, S.; Carroll, A.J.; et al. Mutational landscape and clinical outcome of patients with de novo acute myeloid leukemia and rearrangements involving 11q23/KMT2A. Proc. Natl. Acad. Sci. USA 2020, 117, 26340–26346. [Google Scholar] [CrossRef] [PubMed]
- Yokoyama, A.; Somervaille, T.C.; Smith, K.S.; Rozenblatt-Rosen, O.; Meyerson, M.; Cleary, M.L. The menin tumor suppressor protein is an essential oncogenic cofactor for MLL-associated leukemogenesis. Cell 2005, 123, 207–218. [Google Scholar] [CrossRef] [PubMed]
- Issa, G.C.; Ravandi, F.; DiNardo, C.D.; Jabbour, E.; Kantarjian, H.M.; Andreeff, M. Therapeutic implications of menin inhibition in acute leukemias. Leukemia 2021, 35, 2482–2495. [Google Scholar] [CrossRef] [PubMed]
- Aldoss, I.; Issa, G.C.; Thirman, M.; DiPersio, J.; Arellano, M.; Blachly, J.S.; Mannis, G.N.; Perl, A.; Dickens, D.S.; McMahon, C.M.; et al. Revumenib Monotherapy in Patients with Relapsed/Refractory KMT2Ar Acute Leukemia: Topline Efficacy and Safety Results from the Pivotal Augment-101 Phase 2 Study. Blood 2023, 142 (Suppl. S2), LBA-5. [Google Scholar] [CrossRef]
- Erba, H.P.; Fathi, A.T.; Issa, G.C.; Altman, J.K.; Montesinos, P.; Patnaik, M.M.; Foran, J.M.; De Botton, S.; Baer, M.R.; Schiller, G.J.; et al. Update on a Phase 1/2 First-in-Human Study of the Menin-KMT2A (MLL) Inhibitor Ziftomenib (KO-539) in Patients with Relapsed or Refractory Acute Myeloid Leukemia. Blood 2022, 140 (Suppl. S1), 153–156. [Google Scholar] [CrossRef]
- Kwon, M.C.; Querolle, O.; Dai, X.; Thuring, J.W.; Verhulst, T.; Marien, A.; Goffin, D.; Cai, W.; Keersmaekers, V.; Eyassu, F.; et al. Pharmacological Characterization of JNJ-75276617, a Menin-KMT2A Inhibitor, As Targeted Treatment for KMT2A-Altered and NPM1-Mutant Acute Leukemia. Blood 2022, 140 (Suppl. S1), 5928–5929. [Google Scholar] [CrossRef]
- Stein, E.M.; Garcia-Manero, G.; Rizzieri, D.A.; Tibes, R.; Berdeja, J.G.; Savona, M.R.; Jongen-Lavrenic, M.; Altman, J.K.; Thomson, B.; Blakemore, S.J.; et al. The DOT1L inhibitor pinometostat reduces H3K79 methylation and has modest clinical activity in adult acute leukemia. Blood 2018, 131, 2661–2669. [Google Scholar] [CrossRef] [PubMed]
- Menghrajani, K.; Cai, S.F.; Devlin, S.M.; Armstrong, S.A.; Piekarz, R.; Rudek, M.; Tallman, M.S.; Stein, E.M. A Phase Ib/II Study of the Histone Methyltransferase Inhibitor Pinometostat in Combination with Azacitidine in Patients with 11q23-Rearranged Acute Myeloid Leukemia. Blood 2019, 134 (Suppl. S1), 2655. [Google Scholar] [CrossRef]
- Bourgeois, W.; Cutler, J.A.; Aubrey, B.J.; Wenge, D.; Perner, F.; Martucci, C.; Henrich, J.A.; Klega, K.; Nowak, R.P.; Donovan, K.A.; et al. Mezigdomide is effective alone and in combination with menin inhibition in preclinical models of KMT2A-r and NPM1c AML. Blood 2024, 143, 1513–1527. [Google Scholar] [CrossRef] [PubMed]
- Adema, V.; Colla, S. EZH2 Inhibitors: The Unpacking Revolution. Cancer Res. 2022, 82, 359–361. [Google Scholar] [CrossRef] [PubMed]
- Basheer, F.; Giotopoulos, G.; Meduri, E.; Yun, H.; Mazan, M.; Sasca, D.; Gallipoli, P.; Marando, L.; Gozdecka, M.; Asby, R.; et al. Contrasting requirements during disease evolution identify EZH2 as a therapeutic target in AML. J. Exp. Med. 2019, 216, 966–981. [Google Scholar] [CrossRef] [PubMed]
- Shimony, S.; Bewersdorf, J.P.; Shallis, R.M.; Liu, Y.; Schaefer, E.J.; Zeidan, A.M.; Goldberg, A.D.; Stein, E.M.; Marcucci, G.; Lindsley, R.C.; et al. Hypomethylating agents plus venetoclax compared with intensive induction chemotherapy regimens in molecularly defined secondary AML. Leukemia 2024, 38, 762–768. [Google Scholar] [CrossRef] [PubMed]
- Morschhauser, F.; Tilly, H.; Chaidos, A.; McKay, P.; Phillips, T.; Assouline, S.; Batlevi, C.L.; Campbell, P.; Ribrag, V.; Damaj, G.L.; et al. Tazemetostat for patients with relapsed or refractory follicular lymphoma: An open-label, single-arm, multicentre, phase 2 trial. Lancet Oncol. 2020, 21, 1433–1442. [Google Scholar] [CrossRef]
- Eich, M.L.; Athar, M.; Ferguson, J.E., III; Varambally, S. EZH2-Targeted Therapies in Cancer: Hype or a Reality. Cancer Res. 2020, 80, 5449–5458. [Google Scholar] [CrossRef]
- Porazzi, P.; Petruk, S.; Pagliaroli, L.; De Dominici, M.; Deming, D., II; Puccetti, M.V.; Kushinsky, S.; Kumar, G.; Minieri, V.; Barbieri, E.; et al. Targeting Chemotherapy to Decondensed H3K27me3-Marked Chromatin of AML Cells Enhances Leukemia Suppression. Cancer Res. 2022, 82, 458–471. [Google Scholar] [CrossRef]
- Van Vlierberghe, P.; Patel, J.; Abdel-Wahab, O.; Lobry, C.; Hedvat, C.V.; Balbin, M.; Nicolas, C.; Payer, A.R.; Fernandez, H.F.; Tallman, M.S.; et al. PHF6 mutations in adult acute myeloid leukemia. Leukemia 2011, 25, 130–134. [Google Scholar] [CrossRef]
- Patel, J.P.; Gönen, M.; Figueroa, M.E.; Fernandez, H.; Sun, Z.; Racevskis, J.; Van Vlierberghe, P.; Dolgalev, I.; Thomas, S.; Aminova, O.; et al. Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N. Engl. J. Med. 2012, 366, 1079–1089. [Google Scholar] [CrossRef] [PubMed]
- Lower, K.M.; Turner, G.; Kerr, B.A.; Mathews, K.D.; Shaw, M.A.; Gedeon, Á.K.; Schelley, S.; Eugene Hoyme, H.; White, S.M.; Delatycki, M.B.; et al. Mutations in PHF6 are associated with Börjeson–Forssman–Lehmann syndrome. Nat. Genet. 2002, 32, 661–665. [Google Scholar] [CrossRef]
- Yuan, S.; Gao, M.; Wang, Y.; Lan, Y.; Li, M.; Du, Y.; Li, Y.; Ju, W.; Huang, Y.; Yuan, K.; et al. PHF6 loss reduces leukemia stem cell activity in an acute myeloid leukemia mouse model. Cancer Cell Int. 2024, 24, 66. [Google Scholar] [CrossRef] [PubMed]
- Mousa, N.O.; Gado, M.; Assem, M.M.; Dawood, K.M.; Osman, A. Expression profiling of some Acute Myeloid Leukemia—Associated markers to assess their diagnostic/prognostic potential. Genet. Mol. Biol. 2021, 44, e20190268. [Google Scholar] [CrossRef] [PubMed]
- Hou, S.; Wang, X.; Guo, T.; Lan, Y.; Yuan, S.; Yang, S.; Zhao, F.; Fang, A.; Liu, N.; Yang, W.; et al. PHF6 maintains acute myeloid leukemia via regulating NF-κB signaling pathway. Leukemia 2023, 37, 1626–1637. [Google Scholar] [CrossRef]
- Zhao, Y.; Cai, W.; Hua, Y.; Yang, X.; Zhou, J. The Biological and Clinical Consequences of RNA Splicing Factor U2AF1 Mutation in Myeloid Malignancies. Cancers 2022, 14, 4406. [Google Scholar] [CrossRef]
- Wang, H.; Zhang, N.; Wu, X.; Zheng, X.; Ling, Y.; Gong, Y. Prognostic value of U2AF1 mutant in patients with de novo myelodysplastic syndromes: A meta-analysis. Ann. Hematol. 2019, 98, 2629–2639. [Google Scholar] [CrossRef] [PubMed]
- Bamopoulos, S.A.; Batcha, A.M.N.; Jurinovic, V.; Rothenberg-Thurley, M.; Janke, H.; Ksienzyk, B.; Philippou-Massier, J.; Graf, A.; Krebs, S.; Blum, H.; et al. Clinical presentation and differential splicing of SRSF2, U2AF1 and SF3B1 mutations in patients with acute myeloid leukemia. Leukemia 2020, 34, 2621–2634. [Google Scholar] [CrossRef]
- Qian, J.; Yao, D.-M.; Lin, J.; Qian, W.; Wang, C.-Z.; Chai, H.-Y.; Yang, J.; Li, Y.; Deng, Z.-Q.; Ma, J.-C.; et al. U2AF1 mutations in Chinese patients with acute myeloid leukemia and myelodysplastic syndrome. PLoS ONE 2012, 7, e45760. [Google Scholar] [CrossRef] [PubMed]
- Shirai, C.L.; White, B.S.; Tripathi, M.; Tapia, R.; Ley, J.N.; Ndonwi, M.; Kim, S.; Shao, J.; Carver, A.; Saez, B.; et al. Mutant U2AF1-expressing cells are sensitive to pharmacological modulation of the spliceosome. Nat. Commun. 2017, 8, 14060. [Google Scholar] [CrossRef]
- Bennett, J.; Starczynowski, D.T. IRAK1 and IRAK4 as emerging therapeutic targets in hematologic malignancies. Curr. Opin. Hematol. 2022, 29, 8–19. [Google Scholar] [CrossRef] [PubMed]
- Choudhary, G.S.; Smith, M.A.; Pellagatti, A.; Bhagat, T.D.; Gordon, S.; Pandey, S.; Shah, N.; Aluri, S.; Booher, R.N.; Ramachandra, M.; et al. SF3B1 Mutations Induce Oncogenic IRAK4 Isoforms and Activate Targetable Innate Immune Pathways in MDS and AML. Blood 2019, 134 (Suppl. S1), 4224. [Google Scholar] [CrossRef]
- Smith, M.A.; Choudhary, G.S.; Pellagatti, A.; Choi, K.; Bolanos, L.C.; Bhagat, T.D.; Gordon-Mitchell, S.; Von Ahrens, D.; Pradhan, K.; Steeples, V.; et al. U2AF1 mutations induce oncogenic IRAK4 isoforms and activate innate immune pathways in myeloid malignancies. Nat. Cell Biol. 2019, 21, 640–650. [Google Scholar] [CrossRef] [PubMed]
- Garcia-Manero, G.; Winer, E.S.; DeAngelo, D.J.; Tarantolo, S.; Sallman, D.A.; Dugan, J.; Groepper, S.; Giagounidis, A.; Götze, K.; Metzeler, K.H.; et al. S129: Takeaim Leukemia—A Phase 1/2A Study of the IRAK4 Inhibitor EMAVUSERTIB (CA-4948) as Monotherapy or in Combination with Azacitidine or Venetoclax in Relapsed/Refractory AML or MDS. Hemasphere 2022, 6, 30–31. [Google Scholar] [CrossRef]
- Jiang, M.; Chen, M.; Liu, Q.; Jin, Z.; Yang, X.; Zhang, W. SF3B1 mutations in myelodysplastic syndromes: A potential therapeutic target for modulating the entire disease process. Front. Oncol. 2023, 13, 1116438. [Google Scholar] [CrossRef] [PubMed]
- Fenaux, P.; Platzbecker, U.; Mufti, G.J.; Garcia-Manero, G.; Buckstein, R.; Santini, V.; Díez-Campelo, M.; Finelli, C.; Cazzola, M.; Ilhan, O.; et al. Luspatercept in Patients with Lower-Risk Myelodysplastic Syndromes. N. Engl. J. Med. 2020, 382, 140–151. [Google Scholar] [CrossRef] [PubMed]
- Platzbecker, U.; Della Porta, M.G.; Santini, V.; Zeidan, A.M.; Komrokji, R.S.; Shortt, J.; Valcarcel, D.; Jonasova, A.; Dimicoli-Salazar, S.; Tiong, I.S.; et al. Efficacy and safety of luspatercept versus epoetin alfa in erythropoiesis-stimulating agent-naive, transfusion-dependent, lower-risk myelodysplastic syndromes (COMMANDS): Interim analysis of a phase 3, open-label, randomised controlled trial. Lancet 2023, 402, 373–385. [Google Scholar] [CrossRef]
- FDA Approves Luspatercept-Aamt for Anemia in Adults with MDS. Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-luspatercept-aamt-anemia-adults-mds (accessed on 4 May 2024).
- FDA Approves Luspatercept-Aamt as First-Line Treatment for Adult Patients with Lower-Risk MDS and Anemia Who May Require Transfusions. Available online: https://ascopost.com/news/august-2023/fda-approves-luspatercept-aamt-as-first-line-treatment-for-adult-patients-with-lower-risk-mds-and-anemia-who-may-require-transfusions/ (accessed on 4 May 2024).
- Lindsley, R.C.; Mar, B.G.; Mazzola, E.; Grauman, P.V.; Shareef, S.; Allen, S.L.; Pigneux, A.; Wetzler, M.; Stuart, R.K.; Erba, H.P.; et al. Acute myeloid leukemia ontogeny is defined by distinct somatic mutations. Blood 2015, 125, 1367–1376. [Google Scholar] [CrossRef]
- Steensma, D.P.; Wermke, M.; Klimek, V.M.; Greenberg, P.L.; Font, P.; Komrokji, R.S.; Yang, J.; Brunner, A.M.; Carraway, H.E.; Ades, L.; et al. Phase I First-in-Human Dose Escalation Study of the oral SF3B1 modulator H3B-8800 in myeloid neoplasms. Leukemia 2021, 35, 3542–3550. [Google Scholar] [CrossRef]
- Visconte, V.; Nakashima, M.O.; Rogers, H.J. Mutations in Splicing Factor Genes in Myeloid Malignancies: Significance and Impact on Clinical Features. Cancers 2019, 11, 1844. [Google Scholar] [CrossRef] [PubMed]
- Tatarian, J.; Tupper, N.; Li, P.; Feusier, J.; Abdo, M.; Hyter, S.; Gonzales, P.R.; Zhang, D.; Woodroof, J.; Kelting, S.; et al. Morphologic, immunophenotypic, molecular genetic, and clinical characterization in patients with SRSF2-mutated acute myeloid leukemia. Am. J. Clin. Pathol. 2023, 160, 490–499. [Google Scholar] [CrossRef] [PubMed]
- Zheng, X.; Zhan, Z.; Naren, D.; Li, J.; Yan, T.; Gong, Y. Prognostic value of SRSF2 mutations in patients with de novo myelodysplastic syndromes: A meta-analysis. PLoS ONE 2017, 12, e0185053. [Google Scholar] [CrossRef] [PubMed]
- Prassek, V.V.; Rothenberg-Thurley, M.; Sauerland, M.C.; Herold, T.; Janke, H.; Ksienzyk, B.; Konstandin, N.P.; Goerlich, D.; Krug, U.; Faldum, A.; et al. Genetics of acute myeloid leukemia in the elderly: Mutation spectrum and clinical impact in intensively treated patients aged 75 years or older. Haematologica 2018, 103, 1853–1861. [Google Scholar] [CrossRef]
- van der Werf, I.; Wojtuszkiewicz, A.; Meggendorfer, M.; Hutter, S.; Baer, C.; Heymans, M.; Valk, P.J.M.; Kern, W.; Haferlach, C.; Janssen, J.J.W.M.; et al. Splicing factor gene mutations in acute myeloid leukemia offer additive value if incorporated in current risk classification. Blood Adv. 2021, 5, 3254–3265. [Google Scholar] [CrossRef] [PubMed]
- Berton, G.; Sedaki, B.; Collomb, E.; Benachour, S.; Loschi, M.; Mohty, B.; Saillard, C.; Hicheri, Y.; Rouzaud, C.; Maisano, V.; et al. Poor prognosis of SRSF2 gene mutations in patients treated with VEN-AZA for newly diagnosed acute myeloid leukemia. Leuk. Res. 2024, 141, 107500. [Google Scholar] [CrossRef] [PubMed]
- Grimm, J.; Jentzsch, M.; Bill, M.; Backhaus, D.; Brauer, D.; Küpper, J.; Schulz, J.; Franke, G.; Vucinic, V.; Niederwieser, D.; et al. Clinical implications of SRSF2 mutations in AML patients undergoing allogeneic stem cell transplantation. Am. J. Hematol. 2021, 96, 1287–1294. [Google Scholar] [CrossRef] [PubMed]
- Malaise, M.; Steinbach, D.; Corbacioglu, S. Clinical implications of c-Kit mutations in acute myelogenous leukemia. Curr. Hematol. Malig. Rep. 2009, 4, 77–82. [Google Scholar] [CrossRef] [PubMed]
- Gari, M.; Goodeve, A.; Wilson, G.; Winship, P.; Langabeer, S.; Linch, D.; Vandenberghe, E.; Peake, I.; Reilly, J. c-kit proto-oncogene exon 8 in-frame deletion plus insertion mutations in acute myeloid leukaemia. Br. J. Haematol. 1999, 105, 894–900. [Google Scholar] [CrossRef]
- Ikeda, H.; Kanakura, Y.; Tamaki, T.; Kuriu, A.; Kitayama, H.; Ishikawa, J.; Kanayama, Y.; Yonezawa, T.; Tarui, S.; Griffin, J.D. Expression and functional role of the proto-oncogene c-kit in acute myeloblastic leukemia cells. Blood 1991, 78, 2962–2968. [Google Scholar] [CrossRef] [PubMed]
- Cairoli, R.; Beghini, A.; Grillo, G.; Nadali, G.; Elice, F.; Ripamonti, C.B.; Colapietro, P.; Nichelatti, M.; Pezzetti, L.; Lunghi, M.; et al. Prognostic impact of c-KIT mutations in core binding factor leukemias: An Italian retrospective study. Blood 2006, 107, 3463–3468. [Google Scholar] [CrossRef]
- Ayatollahi, H.; Shajiei, A.; Sadeghian, M.H.; Sheikhi, M.; Yazdandoust, E.; Ghazanfarpour, M.; Shams, S.F.; Shakeri, S. Prognostic Importance of C-KIT Mutations in Core Binding Factor Acute Myeloid Leukemia: A Systematic Review. Hematol./Oncol. Stem Cell Ther. 2017, 10, 1–7. [Google Scholar] [CrossRef]
- Paschka, P.; Marcucci, G.; Ruppert, A.S.; Mrózek, K.; Chen, H.; Kittles, R.A.; Vukosavljevic, T.; Perrotti, D.; Vardiman, J.W.; Carroll, A.J.; et al. Adverse prognostic significance of KIT mutations in adult acute myeloid leukemia with inv(16) and t(8;21): A Cancer and Leukemia Group B Study. J. Clin. Oncol. 2006, 24, 3904–3911. [Google Scholar] [CrossRef] [PubMed]
- Advani, A.S.; Tse, W.; Li, H.; Jia, X.; Elson, P.; Cooper, B.; Ali-Osman, F.; Park, J.; Rao, A.V.; Rizzieri, D.A.; et al. A Phase II Trial of Imatinib Mesylate as Maintenance Therapy for Patients with Newly Diagnosed C-kit-positive Acute Myeloid Leukemia. Clin. Lymphoma Myeloma Leuk. 2021, 21, 113–118. [Google Scholar] [CrossRef]
- Sperling, C.; Schwartz, S.; Büchner, T.; Thiel, E.; Ludwig, W.D. Expression of the stem cell factor receptor C-KIT (CD117) in acute leukemias. Haematologica 1997, 82, 617–621. [Google Scholar] [PubMed]
- Advani, A.S.; Rodriguez, C.; Jin, T.; Jawde, R.A.; Saber, W.; Baz, R.; Kalaycio, M.; Sobecks, R.; Sekeres, M.; Tripp, B.; et al. Increased C-kit intensity is a poor prognostic factor for progression-free and overall survival in patients with newly diagnosed AML. Leuk. Res. 2008, 32, 913–918. [Google Scholar] [CrossRef] [PubMed]
- Kindler, T.; Breitenbuecher, F.; Marx, A.; Beck, J.; Hess, G.; Weinkauf, B.; Duyster, J.; Peschel, C.; Kirkpatrick, C.J.; Theobald, M.; et al. Efficacy and safety of imatinib in adult patients with c-kit–positive acute myeloid leukemia. Blood 2004, 103, 3644–3654. [Google Scholar] [CrossRef]
- Advani, A.S.; Tiu, R.; Saunthararajah, Y.; Maciejewski, J.; Copelan, E.A.; Sobecks, R.; Sekeres, M.A.; Bates, J.; Rush, M.L.; Tripp, B.; et al. A Phase 1 study of imatinib mesylate in combination with cytarabine and daunorubicin for c-kit positive relapsed acute myeloid leukemia. Leuk. Res. 2010, 34, 1622–1626. [Google Scholar] [CrossRef] [PubMed]
- Al-Kali, A.; Tibes, R.; Atherton, P.; Palmer, J.; Alkhateeb, H.B.; Patnaik, M.; Begna, K.; Gangat, N.; Hashmi, S.; He, R.; et al. A phase II study of combination daunorubicin, cytarabine (Ara-c), and nilotinib (TAsigna) (DATA) in patients newly diagnosed with acute myeloid leukemia with KIT expression. Am. J. Hematol. 2023, 98, 472–480. [Google Scholar] [CrossRef]
- Ruhnke, L.; Röllig, C.; Herold, S.; Sauer, T.; Brandts, C.H.; Steffen, B.; Schäfer-Eckart, K.; Krause, S.W.; Hänel, M.; Reichle, A.; et al. Midostaurin in addition to intensive chemotherapy in acute myeloid leukemia with t(8;21) and KIT and/or FLT3- ITD mutations: Results of the SAL MIDOKIT trial. Haematologica 2023, 108, 2520–2525. [Google Scholar] [CrossRef]
- Bowen, D.T.; Frew, M.E.; Hills, R.; Gale, R.E.; Wheatley, K.; Groves, M.J.; Langabeer, S.E.; Kottaridis, P.D.; Moorman, A.V.; Burnett, A.K.; et al. RAS mutation in acute myeloid leukemia is associated with distinct cytogenetic subgroups but does not influence outcome in patients younger than 60 years. Blood 2005, 106, 2113–2119. [Google Scholar] [CrossRef]
- Radich, J.P.; Kopecky, K.J.; Willman, C.L.; Weick, J.; Head, D.; Appelbaum, F.; Collins, S.J. N-ras mutations in adult de novo acute myelogenous leukemia: Prevalence and clinical significance. Blood 1990, 76, 801–807. [Google Scholar] [CrossRef] [PubMed]
- Bacher, U.; Haferlach, T.; Schoch, C.; Kern, W.; Schnittger, S. Implications of NRAS mutations in AML: A study of 2502 patients. Blood 2006, 107, 3847–3853. [Google Scholar] [CrossRef]
- Janssen, J.W.; Steenvoorden, A.C.; Lyons, J.; Anger, B.; Böhlke, J.U.; Bos, J.L.; Seliger, H.; Bartram, C.R. RAS gene mutations in acute and chronic myelocytic leukemias, chronic myeloproliferative disorders, and myelodysplastic syndromes. Proc. Natl. Acad. Sci. USA 1987, 84, 9228–9232. [Google Scholar] [CrossRef]
- Ragon, B.K.; Odenike, O.; Baer, M.R.; Stock, W.; Borthakur, G.; Patel, K.; Han, L.; Chen, H.; Ma, H.; Joseph, L.; et al. Oral MEK 1/2 Inhibitor Trametinib in Combination with AKT Inhibitor GSK2141795 in Patients with Acute Myeloid Leukemia with RAS Mutations: A Phase II Study. Clin. Lymphoma Myeloma Leuk. 2019, 19, 431–440.e13. [Google Scholar] [CrossRef] [PubMed]
- De Melo, M.B.; Lorand-Metze, I.; Lima, C.S.; Saad, S.T.; Costa, F.F. N-ras gene point mutations in Brazilian acute myelogenous leukemia patients correlate with a poor prognosis. Leuk. Lymphoma 1997, 24, 309–317. [Google Scholar] [CrossRef]
- Burgess, M.R.; Hwang, E.; Firestone, A.J.; Huang, T.; Xu, J.; Zuber, J.; Bohin, N.; Wen, T.; Kogan, S.C.; Haigis, K.M.; et al. Preclinical efficacy of MEK inhibition in Nras-mutant AML. Blood 2014, 124, 3947–3955. [Google Scholar] [CrossRef] [PubMed]
- Borthakur, G.; Popplewell, L.; Boyiadzis, M.; Foran, J.M.; Platzbecker, U.; Vey, N.; Roland, W.B.; Olin, R.L.; Raza, A.; Giagounidis, A.; et al. Phase I/II Trial of the MEK1/2 Inhibitor Trametinib (GSK1120212) in Relapsed/Refractory Myeloid Malignancies: Evidence of Activity in Patients with RAS Mutation-Positive Disease. Blood 2012, 120, 677. [Google Scholar] [CrossRef]
- Borthakur, G.; Popplewell, L.; Boyiadzis, M.; Foran, J.; Platzbecker, U.; Vey, N.; Walter, R.B.; Olin, R.; Raza, A.; Giagounidis, A.; et al. Activity of the oral mitogen-activated protein kinase kinase inhibitor trametinib in RAS-mutant relapsed or refractory myeloid malignancies. Cancer 2016, 122, 1871–1879. [Google Scholar] [CrossRef]
- Jain, N.; Curran, E.; Iyengar, N.M.; Diaz-Flores, E.; Kunnavakkam, R.; Popplewell, L.; Kirschbaum, M.H.; Karrison, T.; Erba, H.P.; Green, M.; et al. Phase II study of the oral MEK inhibitor selumetinib in advanced acute myelogenous leukemia: A University of Chicago phase II consortium trial. Clin. Cancer Res. 2014, 20, 490–498. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Shukla, M.; Abdul-Hay, M.; Choi, J.H. Molecular Features and Treatment Paradigms of Acute Myeloid Leukemia. Biomedicines 2024, 12, 1768. https://doi.org/10.3390/biomedicines12081768
Shukla M, Abdul-Hay M, Choi JH. Molecular Features and Treatment Paradigms of Acute Myeloid Leukemia. Biomedicines. 2024; 12(8):1768. https://doi.org/10.3390/biomedicines12081768
Chicago/Turabian StyleShukla, Mihir, Maher Abdul-Hay, and Jun H. Choi. 2024. "Molecular Features and Treatment Paradigms of Acute Myeloid Leukemia" Biomedicines 12, no. 8: 1768. https://doi.org/10.3390/biomedicines12081768
APA StyleShukla, M., Abdul-Hay, M., & Choi, J. H. (2024). Molecular Features and Treatment Paradigms of Acute Myeloid Leukemia. Biomedicines, 12(8), 1768. https://doi.org/10.3390/biomedicines12081768