Intensive Versus Non-intensive Therapy for Patients with Newly Diagnosed Acute Myeloid Leukemia (AML)
DOI:
https://doi.org/10.58931/cht.2025.4273Abstract
Newly approved treatments have increased the options available for patients with acute myeloid leukemia (AML), but have also generated questions concerning the selection of the most appropriate therapy for a given individual (Tables 1 & 2). The trials leading to the approval of these therapies were based on limited genetic data (e.g., cytogenetics, FMS-like tyrosine kinase-3 [FLT3] status) and clinical parameters (e.g., age, comorbidities, therapy, or secondary AML). Data concerning effectiveness or lack of efficacy of a drug or drug regimen in specific AML subgroups is often determined after drug approval. For example, venetoclax (VEN) + azacitidine (AZA) lower intensity therapy (LIT), which is approved for the treatment of patients with newly diagnosed AML deemed ineligible for intensive chemotherapy (IC) or aged >75 years, was found to have limited efficacy in patients with mutated TP53. Despite the regulatory approved indications for VEN‑based LIT, some older and younger patients can be selected for either LIT or IC. Furthermore, with the availability of maintenance therapy after IC16, several important questions have emerged regarding the role of IC in older patients.
No published prospective studies have compared IC with LIT in “fit” patients with newly diagnosed AML to inform treatment choice. Two retrospective propensity score matched real‑world data analyses of outcomes in patients with newly diagnosed AML (irrespective of the genetic profile) who received induction with VEN + AZA or IC, indicated no difference in overall survival (OS). However, one study showed improved complete remission (CR) and/or allogeneic hematopoietic stem cell transplant (alloHCT) rates in favour of IC (60.9% vs. 44.2%, P = 0.006 and 18.1% vs. 8.0%, P = 0.012, respectively). Other single‑centre retrospective studies comparing VEN + AZA with IC have yielded conflicting results. None of these studies provided information concerning the use of oral AZA maintenance therapy. The studies did suggest that outcomes may be dependent on specific genetic abnormalities and/or clinical factors. Currently, several Phase 2 trials are comparing VEN + AZA with IC in adult patients with newly diagnosed AML (NCT04801797, NCT05904106, NCT05554406, NCT05554393).
Here, two case scenarios will be discussed to highlight issues surrounding treatment choice: a) fit individuals who are ≥75 years with newly diagnosed European LeukemiaNet (ELN)-defined favourable-risk AML and b) IC eligible persons who are ≥18 years with newly diagnosed ELN‑defined poor-risk AML, who require alloHCT in first complete remission (CR1) with curative intent.
References
Lancet JE, Uy GL, Cortes JE, Newell LF, Lin TL, Ritchie EK, 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(26):2684-92. DOI: https://doi.org/10.1200/JCO.2017.77.6112
Lancet JE, Uy GL, Newell LF, Lin TL, Ritchie EK, Stuart RK, et al. CPX-351 versus 7+3 cytarabine and daunorubicin chemotherapy in older adults with newly diagnosed high-risk or secondary acute myeloid leukaemia: 5-year results of a randomised, open-label, multicentre, phase 3 trial. Lancet Haematol. 2021;8(7):e481-e91. DOI: https://doi.org/10.1016/S2352-3026(21)00134-4
Castaigne S, Pautas C, Terre C, Raffoux E, Bordessoule D, Bastie JN, et al. Effect of gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid leukaemia (ALFA-0701): a randomised, open-label, phase 3 study. Lancet. 2012;379(9825):1508-16. DOI: https://doi.org/10.1016/S0140-6736(12)60485-1
Lambert J, Pautas C, Terre C, Raffoux E, Turlure P, Caillot D, et al. Gemtuzumab ozogamicin for de novo acute myeloid leukemia: final efficacy and safety updates from the open-label, phase III ALFA-0701 trial. Haematologica. 2019;104(1):113-9. DOI: https://doi.org/10.3324/haematol.2018.188888
Hills RK, Castaigne S, Appelbaum FR, Delaunay J, Petersdorf S, Othus M, et al. Addition of gemtuzumab ozogamicin to induction chemotherapy in adult patients with acute myeloid leukaemia: a meta-analysis of individual patient data from randomised controlled trials. Lancet Oncol. 2014;15(9):986-96. DOI: https://doi.org/10.1016/S1470-2045(14)70281-5
Stone RM, Mandrekar SJ, Sanford BL, Laumann K, Geyer S, Bloomfield CD, et al. Midostaurin plus Chemotherapy for Acute Myeloid Leukemia with a FLT3 Mutation. N Engl J Med. 2017;377(5):454-64. DOI: https://doi.org/10.1056/NEJMoa1614359
Stone RM, Yin J, Mandrekar SJ, Benner A, Saadati M, Galinsky IA, et al. 10 Year Follow-up of CALGB 10603/Ratify: Midostaurin Versus Placebo Plus Intensive Chemotherapy in Newly Diagnosed FLT3 Mutant Acute Myeloid Leukemia Patients Aged 18-60 Years. Blood. 2024;144(Supplement 1):218. DOI: https://doi.org/10.1182/blood-2024-201058
Erba HP, Montesinos P, Kim HJ, Patkowska E, Vrhovac R, Zak P, 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(10388):1571-83. DOI: https://doi.org/10.1016/S0140-6736(23)00464-6
DiNardo CD, Jonas BA, Pullarkat V, Thirman MJ, Garcia JS, Wei AH, et al. Azacitidine and Venetoclax in Previously Untreated Acute Myeloid Leukemia. N Engl J Med. 2020;383(7):617-29. DOI: https://doi.org/10.1056/NEJMoa2012971
Pratz KW, Jonas BA, Pullarkat V, Thirman MJ, Garcia JS, Dohner H, et al. Long-term follow-up of VIALE-A: Venetoclax and azacitidine in chemotherapy-ineligible untreated acute myeloid leukemia. Am J Hematol. 2024;99(4):615-24. DOI: https://doi.org/10.1002/ajh.27246
Wei AH, Montesinos P, Ivanov V, DiNardo CD, Novak J, Laribi K, et al. Venetoclax plus LDAC for newly diagnosed AML ineligible for intensive chemotherapy: a phase 3 randomized placebo-controlled trial. Blood. 2020;135(24):2137-45. DOI: https://doi.org/10.1182/blood.2020004856
Wei AH, Panayiotidis P, Montesinos P, Laribi K, Ivanov V, Kim I, et al. 6-month follow-up of VIALE-C demonstrates improved and durable efficacy in patients with untreated AML ineligible for intensive chemotherapy (141/150). Blood Cancer J. 2021;11(10):163. DOI: https://doi.org/10.1038/s41408-021-00565-6
Montesinos P, Recher C, Vives S, Zarzycka E, Wang J, Bertani G, et al. Ivosidenib and Azacitidine in IDH1-Mutated Acute Myeloid Leukemia. N Engl J Med. 2022;386(16):1519-31. DOI: https://doi.org/10.1056/NEJMoa2117344
Rucker FG, Schlenk RF, Bullinger L, Kayser S, Teleanu V, Kett H, et al. TP53 alterations in acute myeloid leukemia with complex karyotype correlate with specific copy number alterations, monosomal karyotype, and dismal outcome. Blood. 2012;119(9):2114-21. DOI: https://doi.org/10.1182/blood-2011-08-375758
Fleming S, Tsai XC, Morris R, Hou HA, Wei AH. TP53 status and impact on AML prognosis within the ELN 2022 risk classification. Blood. 2023;142(23):2029-33. DOI: https://doi.org/10.1182/blood.2023020855
Wei AH, Dohner H, Pocock C, Montesinos P, Afanasyev B, Dombret H, et al. Oral Azacitidine Maintenance Therapy for Acute Myeloid Leukemia in First Remission. N Engl J Med. 2020;383(26):2526-37. DOI: https://doi.org/10.1056/NEJMoa2004444
Zeidan AM, Pollyea DA, Borate U, Vasconcelos A, Potluri R, Rotter D, et al. Venetoclax plus azacitidine compared with intensive chemotherapy as induction for patients with acute myeloid leukemia: retrospective analysis of an electronic medical record database in the United States. Ann Hematol. 2023;102(4):749-54. DOI: https://doi.org/10.1007/s00277-023-05109-5
Matthews AH, Perl AE, Luger SM, Loren AW, Gill SI, Porter DL, et al. Real-world effectiveness of CPX-351 vs venetoclax and azacitidine in acute myeloid leukemia. Blood Adv. 2022;6(13):3997-4005. DOI: https://doi.org/10.1182/bloodadvances.2022007265
Cherry EM, Abbott D, Amaya M, McMahon C, Schwartz M, Rosser J, et al. Venetoclax and azacitidine compared with induction chemotherapy for newly diagnosed patients with acute myeloid leukemia. Blood Adv. 2021;5(24):5565-73. DOI: https://doi.org/10.1182/bloodadvances.2021005538
Diebold K, Mudd T, Jarodiya J, Parks K, Hardee M, Bachiashvili K, et al. Superior survival with intensive chemotherapy, compared to hypomethylating agent + venetoclax, in patients with intermediate/adverse risk acute myeloid leukemia unable to proceed to transplant. Blood. 2024;144(Supplement 1):2900-1. DOI: https://doi.org/10.1182/blood-2024-204120
Khoury JD, Solary E, Abla O, Akkari Y, Alaggio R, Apperley JF, et al. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Myeloid and Histiocytic/Dendritic Neoplasms. Leukemia. 2022;36(7):1703-19. DOI: https://doi.org/10.1038/s41375-022-01613-1
Arber DA, Orazi A, Hasserjian RP, Borowitz MJ, Calvo KR, Kvasnicka HM, et al. International Consensus Classification of Myeloid Neoplasms and Acute Leukemias: integrating morphologic, clinical, and genomic data. Blood. 2022;140(11):1200-28. DOI: https://doi.org/10.1182/blood.2022015850
Falini B, Brunetti L, Sportoletti P, Martelli MP. NPM1-mutated acute myeloid leukemia: from bench to bedside. Blood. 2020;136(15):1707-21. DOI: https://doi.org/10.1182/blood.2019004226
Dohner H, Wei AH, Appelbaum FR, Craddock C, DiNardo CD, Dombret H, et al. Diagnosis and management of AML in adults: 2022 recommendations from an international expert panel on behalf of the ELN. Blood. 2022;140(12):1345-77. DOI: https://doi.org/10.1182/blood.2022016867
Dohner H, DiNardo CD, Appelbaum FR, Craddock C, Dombret H, Ebert BL, et al. Genetic risk classification for adults with AML receiving less-intensive therapies: the 2024 ELN recommendations. Blood. 2024;144(21):2169-73. DOI: https://doi.org/10.1182/blood.2024025409
Othman J, Potter N, Ivey A, Tazi Y, Papaemmanuil E, Jovanovic J, et al. Molecular, clinical, and therapeutic determinants of outcome in NPM1-mutated AML. Blood. 2024;144(7):714-28. DOI: https://doi.org/10.1182/blood.2024024310
Zale A, Ambinder AJ, Kaduluri VPS. A retrospective analysis of intensive chemotherapy vs. venetoclax/hypomethylating agents for patients aged 60-75 with favorable-risk, NPM1-mutated AML. Blood. 2024;144(Supplement 1):450-1. DOI: https://doi.org/10.1182/blood-2024-210320
Bewersdorf JP, Shimony S, Shallis RM, Liu Y, Berton G, Schaefer EJ, et al. Intensive induction chemotherapy vs hypomethylating agents in combination with venetoclax in NPM1-mutant AML. Blood Adv. 2024;8(18):4845-55. DOI: https://doi.org/10.1182/bloodadvances.2024012858
Juliusson G. Older patients with acute myeloid leukemia benefit from intensive chemotherapy: an update from the Swedish Acute Leukemia Registry. Clin Lymphoma Myeloma Leuk. 2011;11 Suppl 1:S54-9. DOI: https://doi.org/10.1016/j.clml.2011.02.003
Lowenberg B, Ossenkoppele GJ, van Putten W, Schouten HC, Graux C, Ferrant A, et al. High-dose daunorubicin in older patients with acute myeloid leukemia. N Engl J Med. 2009;361(13):1235-48. DOI: https://doi.org/10.1056/NEJMoa0901409
Recher C, Rollig C, Berard E, Bertoli S, Dumas PY, Tavitian S, et al. Long-term survival after intensive chemotherapy or hypomethylating agents in AML patients aged 70 years and older: a large patient data set study from European registries. Leukemia. 2022;36(4):913-22. DOI: https://doi.org/10.1038/s41375-021-01425-9
Schlenk RF, Weber D, Fiedler W, Salih HR, Wulf G, Salwender H, et al. Midostaurin added to chemotherapy and continued single-agent maintenance therapy in acute myeloid leukemia with FLT3-ITD. Blood. 2019;133(8):840-51. DOI: https://doi.org/10.1182/blood-2018-08-869453
Dohner H, Weber D, Krzykalla J, Fiedler W, Kuhn MWM, Schroeder T, et al. Intensive chemotherapy with or without gemtuzumab ozogamicin in patients with NPM1-mutated acute myeloid leukaemia (AMLSG 09-09): a randomised, open-label, multicentre, phase 3 trial. Lancet Haematol. 2023;10(7):e495-e509. DOI: https://doi.org/10.1016/S2352-3026(23)00092-3
Borate U, Welkie RL, Huang Y, Swords RT, Traer E, Stein EM, et al. Demographics, characteristics, survival and outcomes in older, untreated, acute myeloid leukemia patients with NPM1 mutations or KMT2A rearrangements from the Beat AML Master Clinical Trial. Blood. 2024;144(Supplement 1):1564. DOI: https://doi.org/10.1182/blood-2024-211261
Burnett AK, Russell NH, Hills RK, Kell J, Cavenagh J, Kjeldsen L, et al. A randomized comparison of daunorubicin 90 mg/m2 vs 60 mg/m2 in AML induction: results from the UK NCRI AML17 trial in 1206 patients. Blood. 2015;125(25):3878-85. DOI: https://doi.org/10.1182/blood-2015-01-623447
Wei AH, Dohner H, Sayar H, Ravandi F, Montesinos P, Dombret H, et al. Long-term survival with oral azacitidine for patients with acute myeloid leukemia in first remission after chemotherapy: Updated results from the randomized, placebo-controlled, phase 3 QUAZAR AML-001 trial. Am J Hematol. 2023;98(4):E84-E7. DOI: https://doi.org/10.1002/ajh.26847
Roboz GJ, Ravandi F, Wei AH, Dombret H, Thol F, Voso MT, et al. Oral azacitidine prolongs survival of patients with AML in remission independently of measurable residual disease status. Blood. 2022;139(14):2145-55. DOI: https://doi.org/10.1182/blood.2021013404
Dohner H, Wei AH, Roboz GJ, Montesinos P, Thol FR, Ravandi F, et al. Prognostic impact of NPM1 and FLT3 mutations in patients with AML in first remission treated with oral azacitidine. Blood. 2022;140(15):1674-85. DOI: https://doi.org/10.1182/blood.2022016293
Schlenk RF, Paschka P, Krzykalla J, Weber D, Kapp-Schwoerer S, Gaidzik VI, et al. Gemtuzumab Ozogamicin in NPM1-Mutated Acute Myeloid Leukemia: Early Results From the Prospective Randomized AMLSG 09-09 Phase III Study. J Clin Oncol. 2020;38(6):623-32. DOI: https://doi.org/10.1200/JCO.19.01406
Kapp-Schwoerer S, Weber D, Corbacioglu A, Gaidzik VI, Paschka P, Kronke J, et al. Impact of gemtuzumab ozogamicin on MRD and relapse risk in patients with NPM1-mutated AML: results from the AMLSG 09-09 trial. Blood. 2020;136(26):3041-50. DOI: https://doi.org/10.1182/blood.2020005998
Dohner H, Pratz KW, DiNardo CD, Wei AH, Jonas BA, Pullarkat VA, et al. Genetic risk stratification and outcomes among treatment-naive patients with AML treated with venetoclax and azacitidine. Blood. 2024;144(21):2211-22. DOI: https://doi.org/10.1182/blood.2024024944
Pratz KW, Jonas BA, Pullarkat V, Recher C, Schuh AC, Thirman MJ, et al. Measurable Residual Disease Response and Prognosis in Treatment-Naive Acute Myeloid Leukemia With Venetoclax and Azacitidine. J Clin Oncol. 2022;40(8):855-65. DOI: https://doi.org/10.1200/JCO.21.01546
Heiblig M, Requena GA, Tauveron-Jalenques U, Tavernier E, Cornillon J, Carre M, et al. Measurable residual disease (MRD) determinants, kinetics and its impact on survival in patients treated with azacitidine and venetoclax for acute myeloid leukemia in frontline setting : a multicentric study from French Auraml Group. Blood. 2024;144(Supplement 1):846-7. DOI: https://doi.org/10.1182/blood-2024-207764
Terwijn M, van Putten WL, Kelder A, van der Velden VH, Brooimans RA, Pabst T, et al. High prognostic impact of flow cytometric minimal residual disease detection in acute myeloid leukemia: data from the HOVON/SAKK AML 42A study. J Clin Oncol. 2013;31(31):3889-97. DOI: https://doi.org/10.1200/JCO.2012.45.9628
Freeman SD, Virgo P, Couzens S, Grimwade D, Russell N, Hills RK, et al. Prognostic relevance of treatment response measured by flow cytometric residual disease detection in older patients with acute myeloid leukemia. J Clin Oncol. 2013;31(32):4123-31. DOI: https://doi.org/10.1200/JCO.2013.49.1753
Pratz KW, DiNardo CD, Selleslag D, Li J, Yamamoto K, Konopleva M, et al. Postremission cytopenia management in patients with acute myeloid leukemia treated with venetoclax and azacitidine in VIALE-A. Am J Hematol. 2022;97(11):E416-E9. DOI: https://doi.org/10.1002/ajh.26692
Chua CC, Hammond D, Kent A, Tiong IS, Konopleva MY, Pollyea DA, et al. Treatment-free remission after ceasing venetoclax-based therapy in patients with acute myeloid leukemia. Blood Adv. 2022;6(13):3879-83. DOI: https://doi.org/10.1182/bloodadvances.2022007083
Garciaz S, Dumas PY, Bertoli S, Sallman DA, Decroocq J, Belhabri A, et al. Outcomes of acute myeloid leukemia patients who responded to venetoclax and azacitidine and stopped treatment. Am J Hematol. 2024;99(10):1870-6. DOI: https://doi.org/10.1002/ajh.27417
Alibhai SM, Breunis H, Timilshina N, Brignardello-Petersen R, Tomlinson G, Mohamedali H, et al. Quality of life and physical function in adults treated with intensive chemotherapy for acute myeloid leukemia improve over time independent of age. J Geriatr Oncol. 2015;6(4):262-71. DOI: https://doi.org/10.1016/j.jgo.2015.04.002
Timilshina N, Breunis H, Tomlinson GA, Brandwein JM, Buckstein R, Durbano S, et al. Long-term recovery of quality of life and physical function over three years in adult survivors of acute myeloid leukemia after intensive chemotherapy. Leukemia. 2019;33(1):15-25. DOI: https://doi.org/10.1038/s41375-018-0162-5
Roboz GJ, Dohner H, Pocock C, Dombret H, Ravandi F, Jang JH, et al. Oral azacitidine preserves favorable level of fatigue and health-related quality of life for patients with acute myeloid leukemia in remission: results from the phase 3, placebo-controlled QUAZAR AML-001 trial. Haematologica. 2021;106(12):3240-4. DOI: https://doi.org/10.3324/haematol.2021.279174
Dillman RO, Davis RB, Green MR, Weiss RB, Gottlieb AJ, Caplan S, et al. A comparative study of two different doses of cytarabine for acute myeloid leukemia: a phase III trial of Cancer and Leukemia Group B. Blood. 1991;78(10):2520-6. DOI: https://doi.org/10.1182/blood.V78.10.2520.2520
Ivey A, Hills RK, Simpson MA, Jovanovic JV, Gilkes A, Grech A, et al. Assessment of Minimal Residual Disease in Standard-Risk AML. N Engl J Med. 2016;374(5):422-33. DOI: https://doi.org/10.1056/NEJMoa1507471
Heuser M, Freeman SD, Ossenkoppele GJ, Buccisano F, Hourigan CS, Ngai LL, et al. 2021 Update on MRD in acute myeloid leukemia: a consensus document from the European LeukemiaNet MRD Working Party. Blood. 2021;138(26):2753-67. DOI: https://doi.org/10.1182/blood.2021013626
Bataller A, Onate G, Diaz-Beya M, Guijarro F, Garrido A, Vives S, et al. Acute myeloid leukemia with NPM1 mutation and favorable European LeukemiaNet category: outcome after preemptive intervention based on measurable residual disease. Br J Haematol. 2020;191(1):52-61. DOI: https://doi.org/10.1111/bjh.16857
Fenaux P, Jonveaux P, Quiquandon I, Lai JL, Pignon JM, Loucheux-Lefebvre MH, et al. P53 gene mutations in acute myeloid leukemia with 17p monosomy. Blood. 1991;78(7):1652-7. DOI: https://doi.org/10.1182/blood.V78.7.1652.1652
Shahzad M, Amin MK, Daver NG, Shah MV, Hiwase D, Arber DA, et al. What have we learned about TP53-mutated acute myeloid leukemia? Blood Cancer J. 2024;14(1):202. DOI: https://doi.org/10.1038/s41408-024-01186-5
Lachowiez CA, Long N, Saultz J, Gandhi A, Newell LF, Hayes-Lattin B, et al. Comparison and validation of the 2022 European LeukemiaNet guidelines in acute myeloid leukemia. Blood Adv. 2023;7(9):1899-909. DOI: https://doi.org/10.1182/bloodadvances.2022009010
Sargas C, Ayala R, Larrayoz MJ, Chillon MC, Rodriguez-Arboli E, Bilbao C, et al. Comparison of the 2022 and 2017 European LeukemiaNet risk classifications in a real-life cohort of the PETHEMA group. Blood Cancer J. 2023;13(1):77. DOI: https://doi.org/10.1038/s41408-023-00835-5
Daver NG, Iqbal S, Huang J, Renard C, Lin J, Pan Y, et al. Clinical characteristics and overall survival among acute myeloid leukemia patients with TP53 gene mutation or chromosome 17p deletion. Am J Hematol. 2023;98(8):1176-84. DOI: https://doi.org/10.1002/ajh.26941
Short NJ, Montalban-Bravo G, Hwang H, Ning J, Franquiz MJ, Kanagal-Shamanna R, et al. Prognostic and therapeutic impacts of mutant TP53 variant allelic frequency in newly diagnosed acute myeloid leukemia. Blood Adv. 2020;4(22):5681-9. DOI: https://doi.org/10.1182/bloodadvances.2020003120
Badar T, Atallah E, Shallis RM, Goldberg AD, Patel A, Abaza Y, et al. Outcomes of TP53-mutated AML with evolving frontline therapies: Impact of allogeneic stem cell transplantation on survival. Am J Hematol. 2022;97(7):E232-E5. DOI: https://doi.org/10.1002/ajh.26546
Badar T, Shahzad M, Atallah E, Litzow MR, Kharfan-Dabaja MA. Transplant or no transplant for TP53 mutated AML. Oncotarget. 2024;15:674-6. DOI: https://doi.org/10.18632/oncotarget.28652
Badar T, Atallah E, Shallis R, Saliba AN, Patel A, Bewersdorf JP, et al. Survival of TP53-mutated acute myeloid leukemia patients receiving allogeneic stem cell transplantation after first induction or salvage therapy: results from the Consortium on Myeloid Malignancies and Neoplastic Diseases (COMMAND). Leukemia. 2023;37(4):799-806. DOI: https://doi.org/10.1038/s41375-023-01847-7
Eissa Y, Remberger, Jamani K, Chen M, Vasudevan Nampoothiri R, Che A, et al. Outocmes of allogeneic stem cell transplantation in TP53-mutated myeloid malignancies: a mulitcenter Canadian study. EBMT annual meeting 2025 2025:A025.
Nawas MT, Kosuri S. Utility or futility? A contemporary approach to allogeneic hematopoietic cell transplantation for TP53-mutated MDS/AML. Blood Adv. 2024;8(3):553-61. DOI: https://doi.org/10.1182/bloodadvances.2023010417
Grob T, Al Hinai ASA, Sanders MA, Kavelaars FG, Rijken M, Gradowska PL, et al. Molecular characterization of mutant TP53 acute myeloid leukemia and high-risk myelodysplastic syndrome. Blood. 2022;139(15):2347-54. DOI: https://doi.org/10.1182/blood.2021014472
Loke J, Labopin M, Craddock C, Cornelissen JJ, Labussiere-Wallet H, Wagner-Drouet EM, et al. Additional cytogenetic features determine outcome in patients allografted for TP53 mutant acute myeloid leukemia. Cancer. 2022;128(15):2922-31. DOI: https://doi.org/10.1002/cncr.34268
Chan O, Hunter A, Talati C, Sallman DA, Asghari H, Song J, et al. Impact of TP53 gene mutation clearance and conditioning intensity on outcome in MDS or AML patients prior to allogeneic stem cell transplantation. Blood. 2019;134(Supplement 1):149. DOI: https://doi.org/10.1182/blood-2019-131530
Lindsley RC, Gibson CJ, Murdock HM, Stone RM, Cortes JE, Uy GL, et al. Genetic characteristics and outcomes by mutation status in a phase 3 study of CPX-351 versus 7+3 in older adults with newly diagnosed, high-risk/secondary acute myeloid leukemia (AML). Blood. 2019;134:15. DOI: https://doi.org/10.1182/blood-2019-124500
Shimony SO, Murdock H, Keating J, Reilly CR, Tsai HK, Gibson CJ, et al. AML-MR mutations drive the benefit of CPX-351 over 7+3 in the pivotal phase 3 AML trial. Blood. 2024;144(Supplement 1):60-1. DOI: https://doi.org/10.1182/blood-2024-200413
Jen WY, Sasaki K, Ravandi F, Kadia TM, Wang SA, Wang W, et al. Impact of measurable residual disease clearance kinetics in patients with AML undergoing intensive chemotherapy. Blood Adv. 2025;9(4):783-92. DOI: https://doi.org/10.1182/bloodadvances.2024013826
Pollyea DA, Pratz KW, Wei AH, Pullarkat V, Jonas BA, Recher C, et al. Outcomes in Patients with Poor-Risk Cytogenetics with or without TP53 Mutations Treated with Venetoclax and Azacitidine. Clin Cancer Res. 2022;28(24):5272-9. DOI: https://doi.org/10.1158/1078-0432.CCR-22-1183

Published
How to Cite
Issue
Section
License
Copyright (c) 2025 Canadian Hematology Today

This work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.