1
|
Récher C, Dumas P, Bérard E, Tavitian S, Leguay T, Galtier J, Alric C, Bidet A, Delabesse E, Rieu JB, Vial J, Vergez F, Luquet I, Klein E, de Grande A, Sarry A, Zukunft S, Platzbecker U, Müller‐Tidow C, Baldus CD, Bornhäuser M, Serve H, Bertoli S, Pigneux A, Röllig C. Mini-consolidations or intermediate-dose cytarabine for the post-remission therapy of AML patients over 60. A retrospective study from the DATAML and SAL registries. Am J Hematol 2025; 100:23-32. [PMID: 39555737 PMCID: PMC11625969 DOI: 10.1002/ajh.27510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/04/2024] [Accepted: 10/06/2024] [Indexed: 11/19/2024]
Abstract
According to current recommendations, older AML patients in first complete remission (CR) after induction chemotherapy should receive consolidation with intermediate-dose cytarabine (IDAC). However, no study has demonstrated the superiority of IDAC over other regimen. In this retrospective study, we compared the efficacy of mini-consolidations (idarubicin 8 mg/m2 day 1, cytarabine 50 mg/m2/12 h, day 1-5) and IDAC. Inclusion criteria were newly diagnosed AML, age > 60 years, first CR after induction and at least 1 cycle of consolidation. Of the 796 included patients, 322 patients received mini-consolidations and 474 patients received IDAC. Mini-consolidation patients were older, and more often, they had de novo AML and unfavorable risk. The rate of allogeneic transplantation was higher in the IDAC group. The median number of cycles was higher in the mini-consolidation group (4 vs. 2; p < .0001). Median relapse-free survival was 18 months with mini-consolidations and 12 months with IDAC (p = .0064). In multivariate analysis, the risk of relapse or death was significantly higher in the IDAC group (p = .004). Median OS was 36 versus 31 months with mini-consolidations or IDAC, respectively (p = .46). In multivariate analysis, the consolidation regimen had no significant influence on OS (p = .43). In older AML patients, post-remission therapy with mini-consolidations represents an alternative to IDAC.
Collapse
Affiliation(s)
- Christian Récher
- Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de ToulouseUniversité Toulouse III Paul SabatierToulouseFrance
| | - Pierre‐Yves Dumas
- Institut National de la Santé et de la Recherche Médicale, U1312, Service d'Hématologie Clinique et de Thérapie CellulaireUniversité de Bordeaux, CHU BordeauxBordeauxFrance
| | - Emilie Bérard
- Service d'Epidémiologie, CERPOP, Inserm, Centre Hospitalier Universitaire de ToulouseUniversité Toulouse III Paul SabatierToulouseFrance
| | - Suzanne Tavitian
- Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de ToulouseUniversité Toulouse III Paul SabatierToulouseFrance
| | - Thibaut Leguay
- Institut National de la Santé et de la Recherche Médicale, U1312, Service d'Hématologie Clinique et de Thérapie CellulaireUniversité de Bordeaux, CHU BordeauxBordeauxFrance
| | - Jean Galtier
- Institut National de la Santé et de la Recherche Médicale, U1312, Service d'Hématologie Clinique et de Thérapie CellulaireUniversité de Bordeaux, CHU BordeauxBordeauxFrance
| | - Camille Alric
- Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de ToulouseUniversité Toulouse III Paul SabatierToulouseFrance
| | - Audrey Bidet
- Laboratoire d'Hématologie BiologiqueCHU BordeauxBordeauxFrance
| | - Eric Delabesse
- Laboratoire d'Hématologie Biologique, Institut Universitaire du Cancer de Toulouse OncopoleCentre Hospitalier Universitaire de ToulouseToulouseFrance
| | - Jean Baptiste Rieu
- Laboratoire d'Hématologie Biologique, Institut Universitaire du Cancer de Toulouse OncopoleCentre Hospitalier Universitaire de ToulouseToulouseFrance
| | | | - François Vergez
- Laboratoire d'Hématologie Biologique, Institut Universitaire du Cancer de Toulouse OncopoleCentre Hospitalier Universitaire de ToulouseToulouseFrance
| | - Isabelle Luquet
- Laboratoire d'Hématologie Biologique, Institut Universitaire du Cancer de Toulouse OncopoleCentre Hospitalier Universitaire de ToulouseToulouseFrance
| | - Emilie Klein
- Laboratoire d'Hématologie BiologiqueCHU BordeauxBordeauxFrance
| | - Anne‐Charlotte de Grande
- Institut National de la Santé et de la Recherche Médicale, U1312, Service d'Hématologie Clinique et de Thérapie CellulaireUniversité de Bordeaux, CHU BordeauxBordeauxFrance
| | - Audrey Sarry
- Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de ToulouseUniversité Toulouse III Paul SabatierToulouseFrance
| | - Sven Zukunft
- Medizinische Klinik und Poliklinik IUniversitätsklinikum TU DresdenDresdenGermany
| | - Uwe Platzbecker
- Klinik und Poliklinik für Hämatologie, ZelltherapieHämostaseologie und Infektiologie Universitätsklinikum LeipzigLeipzigGermany
| | - Carsten Müller‐Tidow
- Klinik für Hämatologie, Onkologie und RheumatologieUniversitätsklinikum HeidelbergHeidelbergGermany
| | - Claudia D. Baldus
- Klinik für Innere Medizin IIUniversitätsklinikum Schleswig‐HolsteinKielGermany
| | - Martin Bornhäuser
- Medizinische Klinik und Poliklinik IUniversitätsklinikum TU DresdenDresdenGermany
| | - Hubert Serve
- Medizinische Klinik 2Universitätsklinikum FrankfurtFrankfurt/MainGermany
| | - Sarah Bertoli
- Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de ToulouseUniversité Toulouse III Paul SabatierToulouseFrance
| | - Arnaud Pigneux
- Institut National de la Santé et de la Recherche Médicale, U1312, Service d'Hématologie Clinique et de Thérapie CellulaireUniversité de Bordeaux, CHU BordeauxBordeauxFrance
| | - Christoph Röllig
- Medizinische Klinik und Poliklinik IUniversitätsklinikum TU DresdenDresdenGermany
| |
Collapse
|
2
|
Didi I, Alliot JM, Dumas PY, Vergez F, Tavitian S, Largeaud L, Bidet A, Rieu JB, Luquet I, Lechevalier N, Delabesse E, Sarry A, De Grande AC, Bérard E, Pigneux A, Récher C, Simoncini D, Bertoli S. Artificial intelligence-based prediction models for acute myeloid leukemia using real-life data: A DATAML registry study. Leuk Res 2024; 136:107437. [PMID: 38215555 DOI: 10.1016/j.leukres.2024.107437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/19/2023] [Accepted: 01/05/2024] [Indexed: 01/14/2024]
Abstract
We designed artificial intelligence-based prediction models (AIPM) using 52 diagnostic variables from 3687 patients included in the DATAML registry treated with intensive chemotherapy (IC, N = 3030) or azacitidine (AZA, N = 657) for an acute myeloid leukemia (AML). A neural network called multilayer perceptron (MLP) achieved a prediction accuracy for overall survival (OS) of 68.5% and 62.1% in the IC and AZA cohorts, respectively. The Boruta algorithm could select the most important variables for prediction without decreasing accuracy. Thirteen features were retained with this algorithm in the IC cohort: age, cytogenetic risk, white blood cells count, LDH, platelet count, albumin, MPO expression, mean corpuscular volume, CD117 expression, NPM1 mutation, AML status (de novo or secondary), multilineage dysplasia and ASXL1 mutation; and 7 variables in the AZA cohort: blood blasts, serum ferritin, CD56, LDH, hemoglobin, CD13 and disseminated intravascular coagulation (DIC). We believe that AIPM could help hematologists to deal with the huge amount of data available at diagnosis, enabling them to have an OS estimation and guide their treatment choice. Our registry-based AIPM could offer a large real-life dataset with original and exhaustive features and select a low number of diagnostic features with an equivalent accuracy of prediction, more appropriate to routine practice.
Collapse
Affiliation(s)
| | | | - Pierre-Yves Dumas
- Centre Hospitalier Universitaire de Bordeaux, Service d'Hématologie Clinique et de Thérapie Cellulaire, Bordeaux, France; Université de Bordeaux, Bordeaux, France; Institut National de la Santé et de la Recherche Médicale, U1035 Bordeaux, France
| | - François Vergez
- Centre Hospitalo-Universitaire de Toulouse, Institut Universitaire du Cancer Toulouse-Oncopole, Laboratoire d'hématologie, Toulouse, France; Centre de Recherches en Cancérologie de Toulouse, Université Toulouse 3 Paul Sabatier, Toulouse, France
| | - Suzanne Tavitian
- Centre Hospitalo-Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Service d'hématologie, Toulouse, France
| | - Laëtitia Largeaud
- Centre Hospitalo-Universitaire de Toulouse, Institut Universitaire du Cancer Toulouse-Oncopole, Laboratoire d'hématologie, Toulouse, France; Centre de Recherches en Cancérologie de Toulouse, Université Toulouse 3 Paul Sabatier, Toulouse, France
| | - Audrey Bidet
- CHU Bordeaux, Laboratoire d'Hématologie Biologique, F-33000 Bordeaux, France
| | - Jean-Baptiste Rieu
- Centre Hospitalo-Universitaire de Toulouse, Institut Universitaire du Cancer Toulouse-Oncopole, Laboratoire d'hématologie, Toulouse, France
| | - Isabelle Luquet
- Centre Hospitalo-Universitaire de Toulouse, Institut Universitaire du Cancer Toulouse-Oncopole, Laboratoire d'hématologie, Toulouse, France
| | - Nicolas Lechevalier
- CHU Bordeaux, Laboratoire d'Hématologie Biologique, F-33000 Bordeaux, France
| | - Eric Delabesse
- Centre Hospitalo-Universitaire de Toulouse, Institut Universitaire du Cancer Toulouse-Oncopole, Laboratoire d'hématologie, Toulouse, France; Centre de Recherches en Cancérologie de Toulouse, Université Toulouse 3 Paul Sabatier, Toulouse, France
| | - Audrey Sarry
- Centre Hospitalo-Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Service d'hématologie, Toulouse, France
| | - Anne-Charlotte De Grande
- Centre Hospitalier Universitaire de Bordeaux, Service d'Hématologie Clinique et de Thérapie Cellulaire, Bordeaux, France
| | - Emilie Bérard
- Department of Epidemiology, Health Economics and Public Health, UMR 1295 CERPOP, University of Toulouse, INSERM, UPS, Toulouse University Hospital (CHU), Toulouse, France
| | - Arnaud Pigneux
- Centre Hospitalier Universitaire de Bordeaux, Service d'Hématologie Clinique et de Thérapie Cellulaire, Bordeaux, France; Université de Bordeaux, Bordeaux, France
| | - Christian Récher
- Centre de Recherches en Cancérologie de Toulouse, Université Toulouse 3 Paul Sabatier, Toulouse, France; Centre Hospitalo-Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Service d'hématologie, Toulouse, France
| | - David Simoncini
- IRIT UMR 5505-CNRS, Université Toulouse I Capitole, Toulouse, France
| | - Sarah Bertoli
- Centre de Recherches en Cancérologie de Toulouse, Université Toulouse 3 Paul Sabatier, Toulouse, France; Centre Hospitalo-Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Service d'hématologie, Toulouse, France.
| |
Collapse
|
3
|
Röllig C. Improving long-term outcomes with intensive induction chemotherapy for patients with AML. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2023; 2023:175-185. [PMID: 38066853 PMCID: PMC10727094 DOI: 10.1182/hematology.2023000504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Intensive chemotherapy in combination with allogeneic hematopoietic cell transplantation and supportive care can induce long-term remissions in around 50% of acute myeloid leukemia patients eligible for intensive treatment. Several treatment optimization trials helped to refine schedule and dosing of the historic "7 + 3" combination. Together with the addition of novel agents, increased efficacy and tolerability led to improved long-term outcomes. Unsatisfactory outcomes in fit elderly patients and unfavorable genetic subgroups have raised the question of whether less-intensive venetoclax-based approaches may be beneficial as an alternative. Although tempting and worth exploring, this issue will remain controversial until the results of randomized comparisons appear. To date, intensive chemotherapy remains the only evident curative treatment option for long-term disease eradication in a fixed treatment time. With the advent of more novel agents and advances in minimal residual disease (MRD) detection and maintenance approaches, the face of intensive treatment could change in many ways. Several are being explored in clinical trials, such as (1) combinations of more than 1 novel agent with the intensive backbone, (2) head-to-head comparisons of novel agents, (3) replacement or dose reduction of cytotoxic components such as anthracyclines, and (4) MRD-guided escalation and de-escalation strategies. The combination of intensive treatment with individualized tailored innovative strategies will most certainly reduce treatment-related toxicities and increase the chances for long-term remission in the future.
Collapse
Affiliation(s)
- Christoph Röllig
- Department of Internal Medicine I, University Hospital TU Dresden, Dresden, Germany
| |
Collapse
|
4
|
Mounie M, Dumas PY, Liva-Yonnet S, Fabre D, Leguay T, Galtier J, Berard E, Hanta R, Gilleron V, Bertoli S, Pigneux A, Récher C, Costa N. Cost comparison of post-remission strategies in younger and older AML patients in France. Blood Cancer J 2023; 13:100. [PMID: 37380651 DOI: 10.1038/s41408-023-00874-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/05/2023] [Accepted: 06/14/2023] [Indexed: 06/30/2023] Open
Affiliation(s)
- Michael Mounie
- Unité d'Evaluation Médico-Economique, Centre Hospitalier Universitaire (CHU), Toulouse, France.
- Institut National de la Santé et de la Recherche Médicale, U1295, Toulouse, France.
| | - Pierre-Yves Dumas
- CHU Bordeaux, Service d'Hématologie Clinique et de Thérapie Cellulaire, F-33000, Bordeaux, France
- Université de Bordeaux, Bordeaux, France
- Institut National de la Santé et de la Recherche Médicale, U1312, Bordeaux, France
| | - Sandra Liva-Yonnet
- Département d'Information Médicale, Centre Hospitalier Universitaire (CHU), Toulouse, France
| | - Didier Fabre
- Département d'Information Médicale, Centre Hospitalier Universitaire (CHU), Toulouse, France
| | - Thibault Leguay
- CHU Bordeaux, Service d'Hématologie Clinique et de Thérapie Cellulaire, F-33000, Bordeaux, France
| | - Jean Galtier
- CHU Bordeaux, Service d'Hématologie Clinique et de Thérapie Cellulaire, F-33000, Bordeaux, France
| | - Emilie Berard
- Institut National de la Santé et de la Recherche Médicale, U1295, Toulouse, France
| | - Ramaroson Hanta
- Département d'Information Médicale, Centre Hospitalier Universitaire (CHU), Bordeaux, France
| | - Véronique Gilleron
- Département d'Information Médicale, Centre Hospitalier Universitaire (CHU), Bordeaux, France
| | - Sarah Bertoli
- Service d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France
- Université Toulouse III Paul Sabatier, Toulouse, France
- Cancer Research Center of Toulouse, Unité Mixte de Recherche (UMR) 1037 INSERM, ERL5294 Centre National de la Recherche Scientifique, Toulouse, France
| | - Arnaud Pigneux
- Université de Bordeaux, Bordeaux, France
- Institut National de la Santé et de la Recherche Médicale, U1312, Bordeaux, France
| | - Christian Récher
- Service d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France.
- Université Toulouse III Paul Sabatier, Toulouse, France.
- Cancer Research Center of Toulouse, Unité Mixte de Recherche (UMR) 1037 INSERM, ERL5294 Centre National de la Recherche Scientifique, Toulouse, France.
| | - Nadège Costa
- Unité d'Evaluation Médico-Economique, Centre Hospitalier Universitaire (CHU), Toulouse, France
- Institut National de la Santé et de la Recherche Médicale, U1295, Toulouse, France
| |
Collapse
|
5
|
Dumas PY, Pigneux A. [Management of AML in the elderly]. Bull Cancer 2023; 110:424-432. [PMID: 36870810 DOI: 10.1016/j.bulcan.2023.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 02/03/2023] [Indexed: 03/06/2023]
Abstract
Elderly patients with acute myeloid leukemia, ineligible for intensive chemotherapy, have long had a very poor prognosis and have always represented one of the main patient populations included in early phase clinical research trials. In recent years, many molecules have shown very interesting efficacy, often targeted therapies whose indication is based on a specific mutation profile (gilteritinib, ivosidenib), or mutation-independent (venetoclax), but also drugs whose indication is based on a specific biomarker (tamibarotene) or on new generation immunotherapies targeting macrophages (magrolimab) or other immune effectors while targeting leukemic cells resulting in forced immunological synapse (flotetuzumab) or activation of lymphocyte effectors associated with inhibition of the AML cells' stem signature in their microenvironment (cusatuzumab sabatolimab). All of these new strategies are discussed in this review, as well as the challenges of this frail population, which has benefited in recent months from all the major advances in the field, questioning in a second phase the modification of practices in younger patients.
Collapse
Affiliation(s)
- Pierre-Yves Dumas
- CHU de Bordeaux, service d'hématologie clinique et thérapie cellulaire, Inserm U1312, 1, avenue Magellan, 33604 Pessac, France.
| | - Arnaud Pigneux
- CHU de Bordeaux, service d'hématologie clinique et thérapie cellulaire, Inserm U1312, 1, avenue Magellan, 33604 Pessac, France
| |
Collapse
|
6
|
Shimony S, Stahl M, Stone RM. Acute myeloid leukemia: 2023 update on diagnosis, risk-stratification, and management. Am J Hematol 2023; 98:502-526. [PMID: 36594187 DOI: 10.1002/ajh.26822] [Citation(s) in RCA: 154] [Impact Index Per Article: 77.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/01/2022] [Accepted: 12/15/2022] [Indexed: 01/04/2023]
Abstract
DISEASE OVERVIEW Acute myeloid leukemia (AML) is a frequently fatal bone marrow stem cell cancer characterized by unbridled proliferation of malignant marrow stem cells with associated infection, anemia, and bleeding. An improved understanding of pathophysiology, improvements in measurement technology and at least 10 recently approved therapies have led to revamping the diagnostic, prognostic, and therapeutic landscape of AML. DIAGNOSIS One updated and one new classification system were published in 2022, both emphasizing the integration of molecular analysis into daily practice. Differences between the International Consensus Classification and major revisions from the previous 2016 WHO system provide both challenges and opportunities for care and clinical research. RISK ASSESSMENT AND MONITORING The European Leukemia Net 2022 risk classification integrates knowledge from novel molecular findings and recent trial results, as well as emphasizing dynamic risk based on serial measurable residual disease assessment. However, how to leverage our burgeoning ability to measure a small number of potentially malignant myeloid cells into therapeutic decision making is controversial. RISK ADAPTED THERAPY The diagnostic and therapeutic complexity plus the availability of newly approved agents requires a nuanced therapeutic algorithm which should integrate patient goals of care, comorbidities, and disease characteristics including the specific mutational profile of the patient's AML. The framework we suggest only represents the beginning of the discussion.
Collapse
Affiliation(s)
- Shai Shimony
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Rabin Medical Center and Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Maximilian Stahl
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Richard M Stone
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| |
Collapse
|
7
|
How I Treat TP53-Mutated Acute Myeloid Leukemia and Myelodysplastic Syndromes. Cancers (Basel) 2022; 14:cancers14184519. [PMID: 36139679 PMCID: PMC9496940 DOI: 10.3390/cancers14184519] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 12/19/2022] Open
Abstract
TP53-mutated acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) are among the myeloid malignancies with the poorest prognosis. In this review, we analyze the prognosis of these two diseases, focussing particularly on the extent of the mono or biallelic mutation status of TP53 mutation, which is largely correlated with cytogenetic complexity. We discuss the possible/potential improvement in outcome based on recent results obtained with new drugs (especially eprenetapopt and magrolimab). We also focus on the impact of allogeneic hematopoietic stem cell transplantation (aHSCT) including post aHSCT treatment.
Collapse
|
8
|
Duployez N, Largeaud L, Duchmann M, Kim R, Rieunier J, Lambert J, Bidet A, Larcher L, Lemoine J, Delhommeau F, Hirsch P, Fenwarth L, Kosmider O, Decroocq J, Bouvier A, Le Bris Y, Ochmann M, Santagostino A, Adès L, Fenaux P, Thomas X, Micol JB, Gardin C, Itzykson R, Soulier J, Clappier E, Recher C, Preudhomme C, Pigneux A, Dombret H, Delabesse E, Sébert M. Prognostic impact of DDX41 germline mutations in intensively treated acute myeloid leukemia patients: an ALFA-FILO study. Blood 2022; 140:756-768. [PMID: 35443031 PMCID: PMC9389637 DOI: 10.1182/blood.2021015328] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 04/01/2022] [Indexed: 11/20/2022] Open
Abstract
DDX41 germline mutations (DDX41MutGL) are the most common genetic predisposition to myelodysplastic syndrome and acute myeloid leukemia (AML). Recent reports suggest that DDX41MutGL myeloid malignancies could be considered as a distinct entity, even if their specific presentation and outcome remain to be defined. We describe here the clinical and biological features of 191 patients with DDX41MutGL AML. Baseline characteristics and outcome of 86 of these patients, treated with intensive chemotherapy in 5 prospective Acute Leukemia French Association/French Innovative Leukemia Organization trials, were compared with those of 1604 patients with DDX41 wild-type (DDX41WT) AML, representing a prevalence of 5%. Patients with DDX41MutGL AML were mostly male (75%), in their seventh decade, and with low leukocyte count (median, 2 × 109/L), low bone marrow blast infiltration (median, 33%), normal cytogenetics (75%), and few additional somatic mutations (median, 2). A second somatic DDX41 mutation (DDX41MutSom) was found in 82% of patients, and clonal architecture inference suggested that it could be the main driver for AML progression. DDX41MutGL patients displayed higher complete remission rates (94% vs 69%; P < .0001) and longer restricted mean overall survival censored at hematopoietic stem cell transplantation (HSCT) than 2017 European LeukemiaNet intermediate/adverse (Int/Adv) DDX41WT patients (5-year difference in restricted mean survival times, 13.6 months; P < .001). Relapse rates censored at HSCT were lower at 1 year in DDX41MutGL patients (15% vs 44%) but later increased to be similar to Int/Adv DDX41WT patients at 3 years (82% vs 75%). HSCT in first complete remission was associated with prolonged relapse-free survival (hazard ratio, 0.43; 95% confidence interval, 0.21-0.88; P = .02) but not with longer overall survival (hazard ratio, 0.77; 95% confidence interval, 0.35-1.68; P = .5).
Collapse
Affiliation(s)
- Nicolas Duployez
- Hematology Laboratory, Unité 1277-Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER), Centre Hospitalier Universitaire (CHU) de Lille, University of Lille, INSERM, Lille, France
| | - Laëtitia Largeaud
- Hematology Laboratory, CHU de Toulouse-Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Matthieu Duchmann
- Université de Paris, Unité 944/7212-GenCellDi, INSERM and Centre National de la Recherche Scientifique (CNRS), Paris, France
| | - Rathana Kim
- Université de Paris, Unité 944/7212-GenCellDi, INSERM and Centre National de la Recherche Scientifique (CNRS), Paris, France
- Hematology Laboratory, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Julie Rieunier
- Hematology Laboratory, CHU de Toulouse-Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | | | - Audrey Bidet
- Hematology Laboratory, CHU de Bordeaux, Bordeaux, France
| | - Lise Larcher
- Université de Paris, Unité 944/7212-GenCellDi, INSERM and Centre National de la Recherche Scientifique (CNRS), Paris, France
- Hematology Laboratory, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Jean Lemoine
- Hematology Department, Saint Louis Hospital, AP-HP, Paris, France
| | - François Delhommeau
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Laboratoire d'hématologie biologique, Hôpital Saint-Antoine, Paris, France
| | - Pierre Hirsch
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Laboratoire d'hématologie biologique, Hôpital Saint-Antoine, Paris, France
| | - Laurène Fenwarth
- Hematology Laboratory, Unité 1277-Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER), Centre Hospitalier Universitaire (CHU) de Lille, University of Lille, INSERM, Lille, France
| | | | | | - Anne Bouvier
- Hematology Laboratory, CHU Angers, Angers, France
| | - Yannick Le Bris
- Hematology Biology, Nantes University Hospital, Nantes, France
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France
| | | | | | - Lionel Adès
- Université de Paris, Unité 944/7212-GenCellDi, INSERM and Centre National de la Recherche Scientifique (CNRS), Paris, France
- Hematology Department, Saint Louis Hospital, AP-HP, Paris, France
| | - Pierre Fenaux
- Université de Paris, Unité 944/7212-GenCellDi, INSERM and Centre National de la Recherche Scientifique (CNRS), Paris, France
- Hematology Department, Saint Louis Hospital, AP-HP, Paris, France
| | - Xavier Thomas
- Hematology Department, Hospices Civils de Lyon, Lyon-Sud Hospital, Lyon, France
| | - Jean-Baptiste Micol
- Hematology Department, Gustave Roussy Institute, University of Paris-Saclay, Villejuif, France
| | - Claude Gardin
- Hematology Department, Avicenne Hospital, AP-HP, Bobigny, France
- Unité 3518, Saint-Louis Institute for Research, Université de Paris, Paris, France
| | - Raphael Itzykson
- Université de Paris, Unité 944/7212-GenCellDi, INSERM and Centre National de la Recherche Scientifique (CNRS), Paris, France
- Hematology Department, Saint Louis Hospital, AP-HP, Paris, France
| | - Jean Soulier
- Université de Paris, Unité 944/7212-GenCellDi, INSERM and Centre National de la Recherche Scientifique (CNRS), Paris, France
- Hematology Laboratory, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Emmanuelle Clappier
- Université de Paris, Unité 944/7212-GenCellDi, INSERM and Centre National de la Recherche Scientifique (CNRS), Paris, France
- Hematology Laboratory, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Christian Recher
- Service d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Université Toulouse III Paul Sabatier, Toulouse, France; and
| | - Claude Preudhomme
- Hematology Laboratory, Unité 1277-Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER), Centre Hospitalier Universitaire (CHU) de Lille, University of Lille, INSERM, Lille, France
| | - Arnaud Pigneux
- Hematology Department, CHU de Bordeaux, Bordeaux, France
| | - Hervé Dombret
- Hematology Department, Saint Louis Hospital, AP-HP, Paris, France
- Unité 3518, Saint-Louis Institute for Research, Université de Paris, Paris, France
| | - Eric Delabesse
- Hematology Laboratory, CHU de Toulouse-Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Marie Sébert
- Université de Paris, Unité 944/7212-GenCellDi, INSERM and Centre National de la Recherche Scientifique (CNRS), Paris, France
- Hematology Department, Saint Louis Hospital, AP-HP, Paris, France
| |
Collapse
|
9
|
Desprez C, Riou J, Peterlin P, Marchand T, Couturier MA, Villate A, Mear JB, Chevalier P, Guillerm G, Gyan E, Schmidt-Tanguy A, Walter RB, Hunault-Berger M, Orvain C. Comparison of scoring systems evaluating suitability for intensive chemotherapy in adults with acute myeloid leukemia-a Grand Ouest Against Leukemia (GOAL) study. Leukemia 2022; 36:2408-2417. [PMID: 35962057 DOI: 10.1038/s41375-022-01677-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/28/2022] [Accepted: 08/01/2022] [Indexed: 11/09/2022]
Abstract
Several scoring systems have been developed to assess suitability of individual patients for intensive acute myeloid leukemia (AML) therapy. We sought to compare the performance of these scores in a cohort of 428 consecutive adults with AML who received conventional induction chemotherapy in five academic centers in France. All scoring systems identified a subset of patients with increased 28 and 56-day mortality although the prediction accuracy was overall limited with C-statistics of ranging from 0.61 to 0.71 Overall survival (OS) prediction was more limited and restricted to scoring systems that include AML-related parameters. The outcome of 104 patients (24%) considered unsuitable for intensive chemotherapy based on criteria used in recent randomized trials was similar to that of the other 324 patients (28-day mortality, odds ratio [OR] = 1.88, P = 0.2; 56-day mortality, OR = 1.71, P = 0.21; event-free survival, hazard ratio [HR] = 1.08, P = 0.6; OS, HR = 1.25, P = 0.14) with low discrimination (C-statistic: 0.57, 0.56, 0.50, and 0.52 for 28-day, 56-day mortality, EFS, and OS, respectively). Together, our findings indicate that the accuracy of currently available approaches to identify patients at increased risk of early mortality and shortened survival after intensive AML therapy is relatively limited. Caution regarding the use of available scoring systems should be warranted in clinical decision-making.
Collapse
Affiliation(s)
| | - Jérémie Riou
- Univ Angers, CHU Angers, Inserm, CNRS, MINT, SFR ICAT, Angers, France
| | - Pierre Peterlin
- Service d'hématologie clinique, CHU de Nantes, Rennes, France.,Federation hospitalo-universitaire « Grand Ouest against Leukemia », Angers, France
| | - Tony Marchand
- Federation hospitalo-universitaire « Grand Ouest against Leukemia », Angers, France.,Service d'hématologie clinique, CHU de Rennes, Rennes, France.,INSERM U1236, Université Rennes 1, Rennes, France
| | - Marie-Anne Couturier
- Federation hospitalo-universitaire « Grand Ouest against Leukemia », Angers, France.,Service d'Hématologie Clinique, Hôpital Morvan, CHU de Brest, Rennes, France
| | - Alban Villate
- Federation hospitalo-universitaire « Grand Ouest against Leukemia », Angers, France.,Service d'Hématologie et Thérapie Cellulaire, Hôpital Bretonneau, CHRU de Tours, CNRS EMR7001 LNOx, Université de Tours, Rennes, France
| | - Jean-Baptiste Mear
- Federation hospitalo-universitaire « Grand Ouest against Leukemia », Angers, France.,Service d'hématologie clinique, CHU de Rennes, Rennes, France
| | - Patrice Chevalier
- Service d'hématologie clinique, CHU de Nantes, Rennes, France.,Federation hospitalo-universitaire « Grand Ouest against Leukemia », Angers, France
| | - Gaelle Guillerm
- Federation hospitalo-universitaire « Grand Ouest against Leukemia », Angers, France.,Service d'Hématologie Clinique, Hôpital Morvan, CHU de Brest, Rennes, France
| | - Emmanuel Gyan
- Federation hospitalo-universitaire « Grand Ouest against Leukemia », Angers, France.,Service d'Hématologie et Thérapie Cellulaire, Hôpital Bretonneau, CHRU de Tours, CNRS EMR7001 LNOx, Université de Tours, Rennes, France
| | - Aline Schmidt-Tanguy
- Service des maladies du sang, CHU d'Angers, Angers, France.,Federation hospitalo-universitaire « Grand Ouest against Leukemia », Angers, France.,Université d'Angers, Inserm UMR 1307, CNRS UMR 6075, Nantes Université, CRCI2NA, F-49000, Angers, France
| | - Roland B Walter
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.,Department of Medicine, Division of Hematology/Oncology, University of Washington, Seattle, WA, USA.,Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.,Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Mathilde Hunault-Berger
- Service des maladies du sang, CHU d'Angers, Angers, France.,Federation hospitalo-universitaire « Grand Ouest against Leukemia », Angers, France.,Université d'Angers, Inserm UMR 1307, CNRS UMR 6075, Nantes Université, CRCI2NA, F-49000, Angers, France
| | - Corentin Orvain
- Service des maladies du sang, CHU d'Angers, Angers, France. .,Federation hospitalo-universitaire « Grand Ouest against Leukemia », Angers, France. .,Université d'Angers, Inserm UMR 1307, CNRS UMR 6075, Nantes Université, CRCI2NA, F-49000, Angers, France.
| |
Collapse
|
10
|
Devillier R, Forcade E, Garnier A, Guenounou S, Thepot S, Guillerm G, Ceballos P, Hicheri Y, Dumas PY, Peterlin P, Hunault-Berger M, Béné MC, Bouvier A, Chevallier P, Blaise D, Vey N, Pigneux A, Récher C, Huynh A. In-depth time-dependent analysis of the benefit of allo-HSCT for elderly patients with CR1 AML: a FILO study. Blood Adv 2022; 6:1804-1812. [PMID: 34525180 PMCID: PMC8941467 DOI: 10.1182/bloodadvances.2021004435] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 06/23/2021] [Indexed: 11/20/2022] Open
Abstract
The benefit of allogeneic hematopoietic stem cell transplantation (allo-HSCT) for patients with acute myeloid leukemia (AML) aged >60 years remains a matter of debate, notably when performed in first complete remission (CR1). To clarify this issue, the French Innovative Leukemia Organization (FILO) performed a 10-year real-world time-dependent analysis. The study enrolled patients between 60 and 70 years of age with AML in CR1 after intensive chemotherapy with intermediate (IR) or unfavorable (UR) risk according to the European LeukemiaNet (ELN) 2010 classification. The impact of allo-HSCT was analyzed through three models: (1) time-dependent Cox; (2) multistate for dynamic prediction; and (3) super landmark. The study enrolled 369 (73%) IR and 138 (27%) UR patients with AML, 203 of whom received an allo-HSCT. Classical multivariate analysis showed that allo-HSCT significantly improved relapse-free survival (RFS; hazard ratio [HR] [95% confidence interval (CI)], 0.47 [0.35-0.62]; P < .001) and overall survival (OS; HR [95% CI], 0.56 [0.42-0.76]; P < .001), independently of the ELN risk group. With the multistate model, the predicted 5-year probability for IR and UR patients to remain in CR1 without allo-HSCT was 8% and 1%, respectively. Dynamic predictions confirmed that patients without allo-HSCT continue to relapse over time. Finally, the super landmark model showed that allo-HSCT significantly improved RFS (HR [95% CI], 0.47 [0.36-0.62]; P < .001) and OS (HR [95% CI], 0.54 [0.40-0.72]; P < .001). allo-HSCT in CR1 is reported here as significantly improving the outcome of fit older patients with AML. Long-term RFS without allo-HSCT is very low (<10%), supporting allo-HSCT as being the best curative option for these patients.
Collapse
Affiliation(s)
- Raynier Devillier
- Hematology Department, Institut Paoli-Calmettes, Aix Marseille Univ, CNRS, Inserm, CRCM, Marseille, France
| | - Edouard Forcade
- Hematology and Cell Therapy, Centre Hospitalier Universitaire (CHU) Bordeaux, Bordeaux, France
| | | | - Sarah Guenounou
- Service d'Hématologie, Institut Universitaire du Cancer de Toulouse-Oncopole, CHU de Toulouse, Toulouse, France
| | - Sylvian Thepot
- Maladies du sang, FHU-GOAL Univ Angers, Université de Nantes, CHU Angers, Inserm, CRCINA, SFR ICAT, Angers, France
| | - Gaelle Guillerm
- Service d’Hématologie Clinique, CHRU de Brest, Brest, France
| | - Patrice Ceballos
- Clinical Hematology, Montpellier University Hospital, Montpellier, France
| | - Yosr Hicheri
- Hematology Department, Institut Paoli-Calmettes, Aix Marseille Univ, CNRS, Inserm, CRCM, Marseille, France
| | - Pierre-Yves Dumas
- Hematology and Cell Therapy, Centre Hospitalier Universitaire (CHU) Bordeaux, Bordeaux, France
| | | | - Mathilde Hunault-Berger
- Maladies du sang, FHU-GOAL Univ Angers, Université de Nantes, CHU Angers, Inserm, CRCINA, SFR ICAT, Angers, France
| | | | - Anne Bouvier
- Laboratoire d’Hématologie, Centre Hospitalier Universitaire (CHU) Angers, Inserm, CRCINA, Angers, France
| | | | - Didier Blaise
- Hematology Department, Institut Paoli-Calmettes, Aix Marseille Univ, CNRS, Inserm, CRCM, Marseille, France
| | - Norbert Vey
- Hematology Department, Institut Paoli-Calmettes, Aix Marseille Univ, CNRS, Inserm, CRCM, Marseille, France
| | - Arnaud Pigneux
- Hematology and Cell Therapy, Centre Hospitalier Universitaire (CHU) Bordeaux, Bordeaux, France
| | - Christian Récher
- Service d'Hématologie, Institut Universitaire du Cancer de Toulouse-Oncopole, CHU de Toulouse, Toulouse, France
| | - Anne Huynh
- Service d'Hématologie, Institut Universitaire du Cancer de Toulouse-Oncopole, CHU de Toulouse, Toulouse, France
| |
Collapse
|
11
|
Foran JM. Can venetoclax-based therapy replace 7+3 induction in fit older adults with AML? Best Pract Res Clin Haematol 2021; 34:101335. [PMID: 34865692 DOI: 10.1016/j.beha.2021.101335] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Older patients with acute myeloid leukemia (AML) face a dismal prognosis. Although venetoclax-based therapy has led to improved outcomes among unfit older patients with AML, it is not curative and its efficacy and long-term outcomes among fit older patients is unclear. This review provides insights into factors that influence treatment choices among older patients with AML and what we would need to know for venetoclax-based therapy to replace standard intensive 7 + 3 induction therapy.
Collapse
Affiliation(s)
- James M Foran
- Mayo Clinic Cancer Center, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
| |
Collapse
|
12
|
Michaelis LC. Is there an optimal adjunct therapy to traditional cytotoxic induction? Best Pract Res Clin Haematol 2021; 34:101326. [PMID: 34865698 DOI: 10.1016/j.beha.2021.101326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The traditional cytotoxic induction regimen for acute myeloid leukemia (AML) is seven days of standard-dose cytarabine and three days of an anthracycline antibiotic (such as daunorubicin or idarubicin), commonly known as "7 + 3." Many studies have been conducted to find an additional agent that might improve efficacy. Data from select studies has shown, in certain populations, benefit to adding cladribine, clofarabine and lomustine to a traditional backbone. For mutation-based chemotherapy regimens, midostaurin with 7 + 3 is the current standard of care for FLT3-mutant, younger AML patients. As we learn more about the synergism of molecular agents and traditional anti-cancer treatments, we can hopefully develop novel regimens without abandoning some of the benefits of these mutation agnostic historical therapies.
Collapse
|
13
|
Intermediate-dose cytarabine or standard-dose cytarabine plus single-dose anthracycline as post-remission therapy in older patients with acute myeloid leukemia: impact on health care resource consumption and outcomes. Blood Cancer J 2021; 11:180. [PMID: 34775463 PMCID: PMC8590686 DOI: 10.1038/s41408-021-00551-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/12/2021] [Accepted: 09/01/2021] [Indexed: 12/03/2022] Open
|
14
|
Pollyea DA, Barrett J, DiNardo CD, Michaelis LC, Roboz GJ, Le RQ, Norsworthy KJ, de Claro RA, Theoret MR, Pazdur R. Project 2025: Proposals for the Continued Success of Drug Development in Acute Myeloid Leukemia. Clin Cancer Res 2021; 28:816-820. [PMID: 34753779 DOI: 10.1158/1078-0432.ccr-21-2124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/05/2021] [Accepted: 11/03/2021] [Indexed: 11/16/2022]
Abstract
The Food and Drug Administration Oncology Center of Excellence initiated Project 2025 to develop five-year goals in specific areas of oncology drug development. This meeting, in October 2020, brought together a panel of regulators and academic experts in acute myeloid leukemia (AML) to discuss opportunities to maximize the success that has recently occurred in AML drug development. The panel discussed challenges and opportunities in clinical trial design and novel endpoints, and outlined key considerations for drug development to facilitate continued growth in the field.
Collapse
Affiliation(s)
| | - John Barrett
- National Heart, Lung, and Blood Institute, National Institutes of Health
| | - Courtney D DiNardo
- Departments of Leukemia and Stem Cell Transplantation & Cellular Therapy, MD Anderson Cancer Center, University of Texas
| | - Laura C Michaelis
- Department of Medicine, Division of Hematology and Oncology, Medical College of Wisconsin
| | | | - Robert Q Le
- Office of Oncologic Diseases, United States Food and Drug Administration
| | - Kelly J Norsworthy
- Office of Oncologic Diseases, United States Food and Drug Administration
| | - R Angelo de Claro
- Oncology Center of Excellence, United States Food and Drug Administration
| | - Marc R Theoret
- Center for Drug Evaluation and Research, Food and Drug Administration
| | - Richard Pazdur
- Office of Oncology Drug Products, United States Food and Drug Administration
| |
Collapse
|
15
|
Comont T, Nicolau-Travers ML, Bertoli S, Recher C, Vergez F, Treiner E. MAIT cells numbers and frequencies in patients with acute myeloid leukemia at diagnosis: association with cytogenetic profile and gene mutations. Cancer Immunol Immunother 2021; 71:875-887. [PMID: 34477901 DOI: 10.1007/s00262-021-03037-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 08/16/2021] [Indexed: 12/17/2022]
Abstract
Harnessing or monitoring immune cells is actually a major topic in pre-clinical and clinical studies in acute myeloid leukemia (AML). Mucosal-Associated Invariant T cells (MAIT) constitute one of the largest subset of innate-like, cytotoxic T cell subsets in humans. Despite some papers suggesting a role for MAIT cells in cancer, their specific involvement remains unclear, especially in myeloid malignancies. This prospective monocentric study included 216 patients with a newly diagnosed AML. Circulating MAIT cells were quantified by flow cytometry at diagnosis and during intensive chemotherapy. We observed that circulating MAIT cells show a specific decline in AML patients at diagnosis compared to healthy donors. Post-induction monitored patients presented with a drastic drop in MAIT cell numbers, with recovery after one month. We also found correlation between decrease in MAIT cells number and adverse cytogenetic profile. FLT3-ITD and IDH ½ mutations were associated with higher MAIT cell numbers. Patients with high level of activated MAIT cells are under-represented within patients with a favorable cytogenetic profile, and over-represented among patients with IDH1 mutations or bi-allelic CEBPA mutations. We show for the first time that circulating MAIT cells are affected in newly diagnosed AML patients, suggesting a link between MAIT cells and AML progression. Our work fosters new studies to deepen our knowledge about the role of MAIT cells in cancer.
Collapse
Affiliation(s)
- Thibault Comont
- Department of Internal Medicine, IUCT-Oncopole, CHU Toulouse, Toulouse, France
- Laboratory of Hematology, IUCT-Oncopole, CHU Toulouse, Toulouse, France
- Cancer Research Center of Toulouse, Unité Mixte de Recherche (UMR) 1037 INSERM, ERL5294 Centre National de La Recherche Scientifique, Toulouse, France
| | | | - Sarah Bertoli
- Cancer Research Center of Toulouse, Unité Mixte de Recherche (UMR) 1037 INSERM, ERL5294 Centre National de La Recherche Scientifique, Toulouse, France
- Department of Clinical Hematology, IUCT-Oncopole, CHU Toulouse, Toulouse, France
- University Paul Sabatier III, Toulouse, France
| | - Christian Recher
- Cancer Research Center of Toulouse, Unité Mixte de Recherche (UMR) 1037 INSERM, ERL5294 Centre National de La Recherche Scientifique, Toulouse, France
- Department of Clinical Hematology, IUCT-Oncopole, CHU Toulouse, Toulouse, France
- University Paul Sabatier III, Toulouse, France
| | - Francois Vergez
- Laboratory of Hematology, IUCT-Oncopole, CHU Toulouse, Toulouse, France
- Cancer Research Center of Toulouse, Unité Mixte de Recherche (UMR) 1037 INSERM, ERL5294 Centre National de La Recherche Scientifique, Toulouse, France
- University Paul Sabatier III, Toulouse, France
| | - Emmanuel Treiner
- Laboratory of Immunology, CHU Toulouse, Toulouse, France.
- University Paul Sabatier III, Toulouse, France.
- Infinity, Inserm UMR1291, 330 Avenue de Grande Bretagne, 31000, Toulouse, France.
| |
Collapse
|
16
|
Jouzier C, Hamel JF, Dumas PY, Delaunay J, Bonmati C, Guièze R, Hunault M, Banos A, Lioure B, Béné MC, Ianotto JC, Ojeda-Uribe M, Paul F, Bernard M, Jourdan E, Zerazhi H, Vey N, Ifrah N, Recher C, Pigneux A, Cahn JY. Conventional chemotherapy for acute myeloid leukemia in older adults: Impact on nutritional, cognitive, and functional status. Eur J Haematol 2021; 106:859-867. [PMID: 33733520 DOI: 10.1111/ejh.13624] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 11/28/2022]
Abstract
OBJECTIVES The impact of conventional treatment for acute myeloid leukemia (AML) on the nutritional, cognitive, and functional status of elderly patients is seldom studied. This assessment was performed in the context of the LAMSA 2007 trial. METHODS The trial enrolled 424 patients with de novo AML. Among them, 316 benefited from geriatric assessment (GA) including nutritional, cognitive, and functional status and were scored according to Eastern Cooperative Oncology Group (ECOG) and sorror for the prediction of treatment toxicity, morbidity, and mortality. Patients were investigated at diagnosis for three times during follow-up. RESULTS This study showed that AML and its treatment have no impact on cognitive (P = .554) nor functional status (P = .842 for Activity of Daily Living and P = .087 for Instrumental Activities of Daily Living). The nutritional status improved over time (P = .041). None of these three parameters at baseline, associated or not with ECOG and sorror scores, impacted survivals or toxicities. CONCLUSIONS The cognitive, functional, and nutritional status had no impact in this cohort of fit elderly AML patients without unfavorable cytogenetics. The GA tools used provided no additional information compared with ECOG and sorror scores, to predict toxicity, morbidity, or mortality due to intensive chemotherapy.
Collapse
Affiliation(s)
- Claire Jouzier
- Clinical Hematology, Grenoble University Hospital, Grenoble, France
| | | | - Pierre-Yves Dumas
- Clinical Hematology, Bordeaux University Hospital, Bordeaux University, Inserm 1035, Bordeaux, France
| | | | | | - Romain Guièze
- Clinical Hematology, Clermont-Ferrand University Hospital, Clermont-Ferrand, France
| | - Mathilde Hunault
- Clinical Hematology, Angers University Hospital & INSERM U 892/CNRS 6299, Angers, France
| | - Anne Banos
- Clinical Hematology, Cote Basque General Hospital, Bayonne, France
| | - Bruno Lioure
- Clinical Hematology, Strasbourg University Hospital, Strasbourg, France
| | | | | | | | - Franciane Paul
- Clinical Hematology, Montpellier University Hospital, Montpellier, France
| | - Marc Bernard
- Clinical Hematology, Rennes University Hospital, Rennes, France
| | - Eric Jourdan
- Clinical Hematology, Nîmes University Hospital, Nîmes, France
| | - Hacène Zerazhi
- Clinical Hematology, Avignon General Hospital, Avignon, France
| | - Norbert Vey
- Clinical Hematology, Paoli-Calmettes Institute, Marseille, France
| | - Norbert Ifrah
- Clinical Hematology, Angers University Hospital & INSERM U 892/CNRS 6299, Angers, France
| | - Christian Recher
- Clinical Hematology, Toulouse University Hospital, Cancer University Institute Toulouse Oncopole, Paul Sabatier University, Toulouse, France
| | - Arnaud Pigneux
- Clinical Hematology, Bordeaux University Hospital, Bordeaux University, Inserm 1035, Bordeaux, France
| | - Jean-Yves Cahn
- Clinical Hematology, Grenoble University Hospital, Grenoble, France
| | | |
Collapse
|
17
|
Récher C. Clinical Implications of Inflammation in Acute Myeloid Leukemia. Front Oncol 2021; 11:623952. [PMID: 33692956 PMCID: PMC7937902 DOI: 10.3389/fonc.2021.623952] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/04/2021] [Indexed: 02/06/2023] Open
Abstract
Recent advances in the description of the tumor microenvironment of acute myeloid leukemia, including the comprehensive analysis of the leukemic stem cell niche and clonal evolution, indicate that inflammation may play a major role in many aspects of acute myeloid leukemia (AML) such as disease progression, chemoresistance, and myelosuppression. Studies on the mechanisms of resistance to chemotherapy or tyrosine kinase inhibitors along with high-throughput drug screening have underpinned the potential role of glucocorticoids in this disease classically described as steroid-resistant in contrast to acute lymphoblastic leukemia. Moreover, some mutated oncogenes such as RUNX1, NPM1, or SRSF2 transcriptionally modulate cell state in a manner that primes leukemic cells for glucocorticoid sensitivity. In clinical practice, inflammatory markers such as serum ferritin or IL-6 have a strong prognostic impact and may directly affect disease progression, whereas interesting preliminary data suggested that dexamethasone may improve the outcome for AML patients with a high white blood cell count, which paves the way to develop prospective clinical trials that evaluate the role of glucocorticoids in AML.
Collapse
Affiliation(s)
- Christian Récher
- Service d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Université Toulouse III Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| |
Collapse
|
18
|
Largeaud L, Cornillet-Lefebvre P, Hamel JF, Dumas PY, Prade N, Dufrechou S, Plenecassagnes J, Luquet I, Blanchet O, Banos A, Béné MC, Bernard M, Bertoli S, Bonmati C, Fornecker LM, Guièze R, Haddaoui L, Hunault M, Ianotto JC, Jourdan E, Ojeda M, Peterlin P, Vey N, Zerazhi H, Yosr H, Mineur A, Cahn JY, Ifrah N, Récher C, Pigneux A, Delabesse E. Lomustine is beneficial to older AML with ELN2017 adverse risk profile and intermediate karyotype: a FILO study. Leukemia 2020; 35:1291-1300. [PMID: 32943750 DOI: 10.1038/s41375-020-01031-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/23/2020] [Accepted: 08/14/2020] [Indexed: 12/24/2022]
Abstract
We previously reported the benefit of lomustine addition to conventional chemotherapy in older acute myeloid leukemias with nonadverse chromosomal aberrations in the LAM-SA 2007 randomized clinical trial (NCT00590837). A molecular analysis of 52 genes performed in 330 patients included in this trial, 163 patients being treated with lomustine in combination with idarubicin and cytarabine and 167 without lomustine, identified 1088 mutations with an average of 3.3 mutations per patient. NPM1, FLT3, and DNMT3A were the most frequently mutated genes. A putative therapeutic target was identified in 178 patients (54%). Among five molecular classifications analyzed, the ELN2017 risk classification has the stronger association with the clinical evolution. Patients not treated with lomustine have an expected survival prognosis in agreement with this classification regarding the overall and event-free survivals. In strong contrast, lomustine erased the ELN2017 classification prognosis. The benefit of lomustine in nonadverse chromosomal aberrations was restricted to patients with RUNX1, ASXL1, TP53, and FLT3-ITDhigh/NPM1WT mutations in contrast to the intermediate and favorable ELN2017 patients. This post-hoc analysis identified a subgroup of fit elderly AML patients with intermediate cytogenetics and molecular markers who may benefit from lomustine addition to intensive chemotherapy.
Collapse
Affiliation(s)
- Laetitia Largeaud
- Hematology Biology, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopôle, Université Toulouse III Paul Sabatier, Toulouse, France
| | | | | | - Pierre-Yves Dumas
- Clinical Hematology, Bordeaux University Hospital, Bordeaux University, Inserm 1035, Bordeaux, France
| | - Naïs Prade
- Hematology Biology, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopôle, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Stéphanie Dufrechou
- Hematology Biology, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopôle, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Julien Plenecassagnes
- Bioinformatic Department, Claudius Regaud Institute, Institut Universitaire du Cancer de Toulouse Oncopôle, Toulouse, France
| | - Isabelle Luquet
- Hematology Biology, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopôle, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Odile Blanchet
- Hematology Biology, Angers University Hospital, Angers University, Inserm, CRCINA, Angers, France
| | - Anne Banos
- Clinical Hematology, Cote Basque General Hospital, Bayonne, France
| | - Marie C Béné
- Hematology Biology, Nantes University Hospital, Nantes, France
| | - Marc Bernard
- Clinical Hematology, Rennes University Hospital, Rennes, France
| | - Sarah Bertoli
- Clinical Hematology, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopôle, Université Toulouse III Paul Sabatier, Toulouse, France
| | | | | | - Romain Guièze
- Clinical Hematology, Clermont-Ferrand University Hospital, Clermont-Ferrand, France
| | - Lamya Haddaoui
- FILO Tumor Bank, Pitié-Salpêtrière Hospital, Paris, France
| | - Mathilde Hunault
- Clinical Hematology, Angers University Hospital, INSERM U 892/CNRS 6299, Angers, France
| | - Jean Christophe Ianotto
- Clinical Hematology, Institute of Oncology and Hematology, Brest University Hospital, Brest, France
| | - Eric Jourdan
- Clinical Hematology, Nîmes University Hospital, Nîmes, France
| | - Mario Ojeda
- Clinical Hematology, GHRMSA, Hôpital E Muller, Mulhouse, France
| | - Pierre Peterlin
- Clinical Hematology, Nantes University Hospital, Nantes, France
| | - Norbert Vey
- Clinical Hematology, Aix-Marseille University, Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Hacene Zerazhi
- Clinical Hematology, Avignon General Hospital, Avignon, France
| | - Hicheri Yosr
- Clinical Hematology, Aix-Marseille University, Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Marseille, France.,Clinical Hematology, Montpellier University Hospital, Montpellier, France
| | - Ariane Mineur
- Clinical Research Unit, Bordeaux University Hospital, Bordeaux, France.,FILO, Tours University Hospital, Tours, France
| | - Jean-Yves Cahn
- Clinical Hematology, Grenoble University Hospital, Grenoble, France
| | - Norbert Ifrah
- Clinical Hematology, Angers University Hospital, INSERM U 892/CNRS 6299, Angers, France
| | - Christian Récher
- Clinical Hematology, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopôle, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Arnaud Pigneux
- Clinical Hematology, Bordeaux University Hospital, Bordeaux University, Inserm 1035, Bordeaux, France
| | - Eric Delabesse
- Hematology Biology, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopôle, Université Toulouse III Paul Sabatier, Toulouse, France.
| | | |
Collapse
|
19
|
Naoe T. <Editors' Choice> How to improve outcomes of elderly patients with acute myeloid leukemia: era of excitement. NAGOYA JOURNAL OF MEDICAL SCIENCE 2020; 82:151-160. [PMID: 32581396 PMCID: PMC7276402 DOI: 10.18999/nagjms.82.2.151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Among elderly patients with acute myeloid leukemia (AML), especially those who are unfit for intensive chemotherapy, a policy of reduced-intensity chemotherapy or conservative observation has been chosen, resulting in unmet medical needs. Clinical trials using anticancer drugs including antimetabolites or drugs targeted to cell cycle-related molecules failed to show superiority over conventional treatments. Recently, drugs targeted to Bcl-2, SMO, FLT3, and IDH1/2 have been shown to prolong overall survival alone or in combination with reduced-intensity chemotherapy. These treatments are likely to reshape the therapeutic landscape of AML, which will be personalized for individual patients based on leukemia genetics.
Collapse
Affiliation(s)
- Tomoki Naoe
- National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| |
Collapse
|
20
|
CD34 +CD38 -CD123 + Leukemic Stem Cell Frequency Predicts Outcome in Older Acute Myeloid Leukemia Patients Treated by Intensive Chemotherapy but Not Hypomethylating Agents. Cancers (Basel) 2020; 12:cancers12051174. [PMID: 32384744 PMCID: PMC7281486 DOI: 10.3390/cancers12051174] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 04/30/2020] [Accepted: 05/05/2020] [Indexed: 12/02/2022] Open
Abstract
The prognostic impact of immunophenotypic CD34+CD38−CD123+ leukemic stem cell (iLSC) frequency at diagnosis has been demonstrated in younger patients treated by intensive chemotherapy, however, this is less clear in older patients. Furthermore, the impact of iLSC in patients treated by hypomethylating agents is unknown. In this single-center study, we prospectively assessed the CD34+CD38−CD123+ iLSC frequency at diagnosis in acute myeloid leukemia (AML) patients aged 60 years or older. In a cohort of 444 patients, the median percentage of iLSC at diagnosis was 4.3%. Significant differences were found between treatment groups with a lower median in the intensive chemotherapy group (0.6%) compared to hypomethylating agents (8.0%) or supportive care (11.1%) (p <0.0001). In the intensive chemotherapy group, the median overall survival was 34.5 months in patients with iLSC ≤0.10% and 14.6 months in patients with >0.10% (p = 0.031). In the multivariate analyses of this group, iLSC frequency was significantly and independently associated with the incidence of relapse, event-free, relapse-free, and overall survival. However, iLSC frequency had no prognostic impact on patients treated by hypomethylating agents. Thus, the iLSC frequency at diagnosis is an independent prognostic factor in older acute myeloid patients treated by intensive chemotherapy but not hypomethylating agents.
Collapse
|
21
|
Vey N. Low-intensity regimens versus standard-intensity induction strategies in acute myeloid leukemia. Ther Adv Hematol 2020; 11:2040620720913010. [PMID: 32215195 PMCID: PMC7081460 DOI: 10.1177/2040620720913010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 02/12/2020] [Indexed: 12/11/2022] Open
Abstract
Treatment options for elderly patients with acute myeloid leukemia (AML) remain limited. In this age group, AML is frequently associated with poor-risk features, while patients’ present comorbidities and reduced functional reserves. As such, intensive chemotherapy (ICT) is frequently too toxic or ineffective in elderly patients and is restricted to a select minority, though it is standard therapy for the youngest and fittest patients or for those belonging to either the favorable or intermediate-risk groups. The use of hypomethylating agents represent an effective alternative for patients who are unfit for ICT, yet the results remain unsatisfactory. In recent years, prognostic scores were developed that include geriatric assessment tools and improved risk-stratification. In addition, several effective new drugs have emerged. The combination of these drugs with hypomethylating agents or low-dose cytarabine has produced encouraging preliminary results that may change standard practices and offer an alternative to the dilemma of ICT versus low-intensity therapies.
Collapse
Affiliation(s)
- Norbert Vey
- Institut Paoli-Calmettes, 232 Boulevard de Sainte Marguerite, Marseille, 13009, France
| |
Collapse
|
22
|
Post-remission therapy of adults aged 60 and older with acute myeloid leukemia in first complete remission: role of treatment intensity on the outcome. Ann Hematol 2020; 99:773-780. [PMID: 32088745 DOI: 10.1007/s00277-020-03922-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 01/14/2020] [Indexed: 10/24/2022]
Abstract
Although complete remission (CR) is achieved in 50 to 70% of older fit patients with acute myeloid leukemia (AML), consolidation therapy in this age group remains challenging. In this retrospective study, we aimed to compare outcome in elderly patients treated with different post-remission modalities, including allogenic and autologous hematopoietic stem cell transplantation (HSCT), intensive chemotherapy, and standard-dose chemotherapy (repeated 1 + 5 regimen). We collected data of 441 patients ≥ 60 years in first CR from a single institution. Median age was 67 years. Sixty-one (14%) patients received allo-HSCT, 51 (12%) auto-HSCT, 70 (16%) intensive chemotherapy with intermediate- or high-dose cytarabine (I/HDAC), and 190 (43%) 1 + 5 regimen. Median follow-up was 6.5 years. In multivariate analysis, allo-HSCT, cytogenetics, and PS had a significant impact on OS and LFS. In spite of a more favorable-risk profile, the patients who received I/HDAC had no significantly better LFS as compared with patients treated with 1 + 5 (median LFS 8.8 months vs 10.6 months, p = 0.96). In transplanted patients, median LFS was 13.3 months for auto-HSCT and 25.8 months for allo-HSCT. Pre-transplant chemotherapy with I/HDAC had no effect on the outcome. Toxicity was significantly increased for both transplanted and non-transplanted patients treated with I/HDAC, with more units of blood and platelet transfusion and more time spent in hospitalization, but no higher non-relapse mortality. This study shows that post-remission chemotherapy intensification is not associated with significantly better outcome as compared with standard-dose chemotherapy in elderly patients for whom, overall results remain disappointing.
Collapse
|
23
|
Ossenkoppele GJ, Breems DA, Stuessi G, van Norden Y, Bargetzi M, Biemond BJ, A von dem Borne P, Chalandon Y, Cloos J, Deeren D, Fehr M, Gjertsen B, Graux C, Huls G, Janssen JJJW, Jaspers A, Jongen-Lavrencic M, de Jongh E, Klein SK, van der Klift M, van Marwijk Kooy M, Maertens J, Michaux L, van der Poel MWM, van Rhenen A, Tick L, Valk P, Vekemans MC, van der Velden WJFM, de Weerdt O, Pabst T, Manz M, Löwenberg B. Lenalidomide added to standard intensive treatment for older patients with AML and high-risk MDS. Leukemia 2020; 34:1751-1759. [PMID: 32020044 DOI: 10.1038/s41375-020-0725-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 12/24/2019] [Accepted: 01/22/2020] [Indexed: 11/09/2022]
Abstract
More effective treatment modalities are urgently needed in patients with acute myeloid leukemia (AML) of older age. We hypothesized that adding lenalidomide to intensive standard chemotherapy might improve their outcome. After establishing a safe lenalidomide, dose elderly patients with AML were randomly assigned in this randomized Phase 2 study (n = 222) to receive standard chemotherapy ("3 + 7") with or without lenalidomide at a dose of 20 mg/day 1-21. In the second cycle, patients received cytarabine 1000 mg/m2 twice daily on days 1-6 with or without lenalidomide (20 mg/day 1-21). The CR/CRi rates in the two arms were not different (69 vs. 66%). Event-free survival (EFS) at 36 months was 19% for the standard arm versus 21% for the lenalidomide arm and overall survival (OS) 35% vs. 30%, respectively. The frequencies and grade of adverse events were not significantly different between the treatment arms. Cardiovascular toxicities were rare and equally distributed between the arms. The results of the present study show that the addition of lenalidomide to standard remission induction chemotherapy does not improve the therapeutic outcome of older AML patients. This trial is registered as number NTR2294 in The NederlandsTrial Register (www.trialregister.nl).
Collapse
Affiliation(s)
- G J Ossenkoppele
- Amsterdam University Medical Cente, location VUMC, Amsterdam, Netherlands.
| | | | - G Stuessi
- Bellinzona-IOSI, Bellinzona, Switzerland
| | - Y van Norden
- HOVON Data Center, Erasmus MC- Department of Hematology, Rotterdam, The Netherlands
| | - M Bargetzi
- Aarau- Kantonsspital, Aarau, Switzerland
| | - B J Biemond
- Amsterdam University Medical Center, location AMC, Amsterdam, Netherlands
| | | | - Y Chalandon
- University Hospital and University of Geneva, Genève, Switzerland
| | - J Cloos
- Amsterdam University Medical Cente, location VUMC, Amsterdam, Netherlands
| | - D Deeren
- Roeselare-AZ Delta, Roeselare, Belgium
| | - M Fehr
- St Gallen-Kantonnsspital, St. Gallen, Switzerland
| | - B Gjertsen
- Haukeland University Hospital, Bergen (N), Norway
| | - C Graux
- Yvoir-MontGodinne, Yvoir, Belgium
| | - G Huls
- University Medical Center, Groningen, Netherlands
| | - J J J W Janssen
- Amsterdam University Medical Cente, location VUMC, Amsterdam, Netherlands
| | - A Jaspers
- Hôpital Citadelle, Liège (B), Belgium
| | | | | | - S K Klein
- Meander Medical Center, Amersfoort, Netherlands
| | | | | | - J Maertens
- Hospital Gasthuisberg, Leuven (B), Belgium
| | - L Michaux
- Center for Human Genetics, KU Leuven and University Hospitals Leuven, Leuven, Belgium
| | | | | | - L Tick
- MaximaMC Eindhoven, Eindhoven, Netherlands
| | - P Valk
- Hôpital Citadelle, Liège (B), Belgium
| | | | | | - O de Weerdt
- St Antonius Hospital, Nieuwegein, Netherlands
| | - T Pabst
- Department of Oncology, University Hospital, Inselspital and University of Bern, Bern, Switzerland
| | - M Manz
- University Hospital, Zurich, Switzerland
| | | | | |
Collapse
|
24
|
Abstract
In recent years, several drugs-including midostaurin, gilteritinib, and gemtuzumab ozogamicin, to name a few-have been approved or reapproved in the United States to treat patients with acute myeloid leukemia (AML). Yet survival rates for younger patients had improved with chemotherapy alone even before the approvals of these new agents. This begs the question whether the new therapies will actually have a positive impact on survival. The 5-year survival rate for older patients has also risen, again without the addition of these new agents. The challenge will be to incorporate new therapies and use them where they will have the greatest impact-major work for clinicians and researchers alike.
Collapse
Affiliation(s)
- Jacob M Rowe
- Technion, Israel Institute of Technology, Haifa, Israel; Department of Hematology, Rambam Health Care Campus, Haifa, Israel; Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Department of Hematology, Shaare Zedek Medical Center, 12 Shmuel Bait St, Jerusalem, IL, 9103102, Israel.
| |
Collapse
|
25
|
Derman BA, Larson RA. Post-remission therapy in acute myeloid leukemia: Are we ready for an individualized approach? Best Pract Res Clin Haematol 2019; 32:101102. [PMID: 31779969 DOI: 10.1016/j.beha.2019.101102] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Recent advances in remission induction treatment strategies for acute myeloid leukemia (AML) have improved the rates of complete remission (CR) and overall survival (OS), owing to a concerted effort to tailor therapies toward specific AML subtypes. However, without effective post-remission therapy, most patients will relapse. The extent to which post-remission therapies is individualized in the current paradigm is quite varied. Core binding factor (CBF) AML is typically treated with post-remission high-dose cytarabine (HiDAC) without allogeneic hematopoietic stem cell transplantation (HSCT), whereas those with intermediate or adverse-risk cytogenetics are treated with post-remission cytarabine followed by allogeneic HSCT in CR1 when feasible. A lack of clarity regarding the proper dosing of post-remission cytarabine has made consensus building on dosing and schedule a challenge. CBF AML benefits most from high-dose cytarabine (HiDAC), and dasatinib appears promising as an adjunct for those for KIT-mutated CBF AML. Other than series using CPX-351 or lomustine in older adults, multiagent chemotherapy approaches have resulted in excess toxicity without a survival benefit. Neither hypomethylating agents nor gemtuzumab ozogamicin have shown a material OS benefit. Targeted agents such as FLT3 inhibitors and IDH1/IDH2 inhibitors show potential for the patients who harbor these druggable targets, but few data are available. Many studies evaluating post-remission strategies to target AML in the MRD-positive state are already underway, and these remain a promising area of investigation.
Collapse
Affiliation(s)
- Benjamin A Derman
- Section of Hematology Oncology, Department of Medicine and Comprehensive Cancer Center, University of Chicago, Chicago, IL, USA.
| | - Richard A Larson
- Section of Hematology Oncology, Department of Medicine and Comprehensive Cancer Center, University of Chicago, Chicago, IL, USA
| |
Collapse
|
26
|
Garciaz S, N'guyen Dasi L, Finetti P, Chevalier C, Vernerey J, Poplineau M, Platet N, Audebert S, Pophillat M, Camoin L, Bertucci F, Calmels B, Récher C, Birnbaum D, Chabannon C, Vey N, Duprez E. Epigenetic down-regulation of the HIST1 locus predicts better prognosis in acute myeloid leukemia with NPM1 mutation. Clin Epigenetics 2019; 11:141. [PMID: 31606046 PMCID: PMC6790061 DOI: 10.1186/s13148-019-0738-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 09/05/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The epigenetic machinery is frequently altered in acute myeloid leukemia. Focusing on cytogenetically normal (CN) AML, we previously described an abnormal H3K27me3 enrichment covering 70 kb on the HIST1 cluster (6.p22) in CN-AML patient blasts. Here, we further investigate the molecular, functional, and prognosis significance of this epigenetic alteration named H3K27me3 HIST1 in NPM1-mutated (NPM1mut) CN-AML. RESULTS We found that three quarter of the NPM1mut CN-AML patients were H3K27me3 HIST1high. H3K27me3 HIST1high group of patients was associated with a favorable outcome independently of known molecular risk factors. In gene expression profiling, the H3K27me3 HIST1high mark was associated with lower expression of the histone genes HIST1H1D, HIST1H2BG, HIST1H2AE, and HIST1H3F and an upregulation of genes involved in myelomonocytic differentiation. Mass spectrometry analyses confirmed that the linker histone protein H1d, but not the other histone H1 subtypes, was downregulated in the H3K27me3 HIST1high group of patients. H1d knockdown primed ATRA-mediated differentiation of OCI-AML3 and U937 AML cell lines, as assessed on CD11b/CD11c markers, morphological and gene expression analyses. CONCLUSIONS Our data suggest that NPM1mut AML prognosis depends on the epigenetic silencing of the HIST1 cluster and that, among the H3K27me3 silenced histone genes, HIST1H1D plays a role in AML blast differentiation.
Collapse
Affiliation(s)
- Sylvain Garciaz
- Epigenetic Factors in Normal and Malignant Hematopoiesis Team, Aix Marseille University, CNRS, Inserm, Institut Paoli-Calmettes, CRCM, 27 Boulevard Lei Roure, 13273, Marseille Cedex 09, France
| | - Lia N'guyen Dasi
- Epigenetic Factors in Normal and Malignant Hematopoiesis Team, Aix Marseille University, CNRS, Inserm, Institut Paoli-Calmettes, CRCM, 27 Boulevard Lei Roure, 13273, Marseille Cedex 09, France
| | - Pascal Finetti
- Predictive Oncology Laboratory, CRCM, Inserm, U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille University, Marseille, France
| | - Christine Chevalier
- Epigenetic Factors in Normal and Malignant Hematopoiesis Team, Aix Marseille University, CNRS, Inserm, Institut Paoli-Calmettes, CRCM, 27 Boulevard Lei Roure, 13273, Marseille Cedex 09, France.,Institut Pasteur, G5 Chromatin and Infection, Paris, France
| | - Julien Vernerey
- Epigenetic Factors in Normal and Malignant Hematopoiesis Team, Aix Marseille University, CNRS, Inserm, Institut Paoli-Calmettes, CRCM, 27 Boulevard Lei Roure, 13273, Marseille Cedex 09, France
| | - Mathilde Poplineau
- Epigenetic Factors in Normal and Malignant Hematopoiesis Team, Aix Marseille University, CNRS, Inserm, Institut Paoli-Calmettes, CRCM, 27 Boulevard Lei Roure, 13273, Marseille Cedex 09, France
| | - Nadine Platet
- Epigenetic Factors in Normal and Malignant Hematopoiesis Team, Aix Marseille University, CNRS, Inserm, Institut Paoli-Calmettes, CRCM, 27 Boulevard Lei Roure, 13273, Marseille Cedex 09, France
| | - Stéphane Audebert
- Aix-Marseille University, Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Marseille Protéomique, Marseille, France
| | - Matthieu Pophillat
- Aix-Marseille University, Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Marseille Protéomique, Marseille, France
| | - Luc Camoin
- Aix-Marseille University, Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Marseille Protéomique, Marseille, France
| | - François Bertucci
- Predictive Oncology Laboratory, CRCM, Inserm, U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille University, Marseille, France
| | - Boris Calmels
- Epigenetic Factors in Normal and Malignant Hematopoiesis Team, Aix Marseille University, CNRS, Inserm, Institut Paoli-Calmettes, CRCM, 27 Boulevard Lei Roure, 13273, Marseille Cedex 09, France.,Aix-Marseille University, Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Centre d'Investigations Cliniques en Biothérapies, Marseille, France
| | - Christian Récher
- Service d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France Université Toulouse III Paul Sabatier, Cancer Research Center of Toulouse, UMR1037-INSERM, ERL5294 CNRS, Toulouse, France
| | - Daniel Birnbaum
- Predictive Oncology Laboratory, CRCM, Inserm, U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille University, Marseille, France
| | - Christian Chabannon
- Epigenetic Factors in Normal and Malignant Hematopoiesis Team, Aix Marseille University, CNRS, Inserm, Institut Paoli-Calmettes, CRCM, 27 Boulevard Lei Roure, 13273, Marseille Cedex 09, France.,Aix-Marseille University, Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Centre d'Investigations Cliniques en Biothérapies, Marseille, France
| | - Norbert Vey
- Aix-Marseille University, Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Estelle Duprez
- Epigenetic Factors in Normal and Malignant Hematopoiesis Team, Aix Marseille University, CNRS, Inserm, Institut Paoli-Calmettes, CRCM, 27 Boulevard Lei Roure, 13273, Marseille Cedex 09, France.
| |
Collapse
|
27
|
Abstract
For decades, the standard induction for patients with acute myeloid leukemia (AML) has been the combination of cytarabine with anthracycline (7 + 3 regimen). In August 2017 the US FDA approved CPX-351 (vyxeos), a liposomal formulation of cytarabine and daunorubicin at a fixed 5:1 molar ratio, for the treatment of adults with newly diagnosed AML with myelodysplasia-related changes (AML-MRC) and therapy-related AML (t-AML). This is the first approved treatment specifically for patients with this subgroup of AML. The approval was based on findings from a multicenter, randomized, open-label, phase III study of CPX-351 Versus 7 + 3 in patients 60-75 years old with newly diagnosed AML-MRC or t-AML. In this study CPX-351 had a higher median OS than 7 + 3 (9.56 vs 5.95 months, HR 0.69; 95% CI: 0.52 to 0.90, p = 0.005). In this profile, we review preclinical and clinical data, and discuss limitations and future directions with CPX-351 use in AML.
Collapse
Affiliation(s)
- Mansour Alfayez
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hagop Kantarjian
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Tapan Kadia
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Farhad Ravandi-Kashani
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Naval Daver
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
28
|
Bertoli S, Tavitian S, Bories P, Luquet I, Delabesse E, Comont T, Sarry A, Huguet F, Bérard E, Récher C. Outcome of patients aged 60-75 years with newly diagnosed secondary acute myeloid leukemia: A single-institution experience. Cancer Med 2019; 8:3846-3854. [PMID: 31173485 PMCID: PMC6639188 DOI: 10.1002/cam4.2020] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/09/2019] [Accepted: 01/16/2019] [Indexed: 12/19/2022] Open
Abstract
A recent phase 3 trial showed that outcome of older patients with secondary acute myeloid leukemia (AML) may be improved by a liposomal encapsulation of cytarabine and daunorubicin (CPX‐351). This phase 3 study represents a unique example of prospective data in this rare subgroup providing basis for comparison with real life data. Here, we retrospectively assessed characteristics and outcome of patients aged 60‐75 years with secondary or therapy‐related AML in real life. Out of 218 patients that fulfilled CPX‐351 study criteria, 181 patients (83.0%) received antileukemic treatment either intensive chemotherapy (n = 121) or hypomethylating agents (HMA, n = 60). As compared with patients treated by chemotherapy, HMA‐treated patients were older, had lower WBC, more often AML with antecedent myelodysplastic syndrome and adverse cytogenetic risk. In chemotherapy‐treated patients, the complete response rate was 69%, median overall survival (OS) was 11 months whereas 3‐year and 5‐year OS was 21% and 17%, respectively. In HMA‐treated patients, the complete response rate was 15%, median OS was 11 months whereas 3‐year and 5‐year OS was 15% and 2%, respectively. In conclusion, although outcome of older patients with high‐risk AML is very poor, a significant proportion of patients treated by standard intensive chemotherapy but not HMA are long‐term survivors.
Collapse
Affiliation(s)
- Sarah Bertoli
- Service d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France.,Université Toulouse III Paul Sabatier, Toulouse, France.,Cancer Research Center of Toulouse, UMR1037-INSERM, ERL5294 CNRS, Toulouse, France
| | - Suzanne Tavitian
- Service d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France
| | - Pierre Bories
- Réseau Onco-occitanie, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France
| | - Isabelle Luquet
- Laboratoire d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France
| | - Eric Delabesse
- Université Toulouse III Paul Sabatier, Toulouse, France.,Cancer Research Center of Toulouse, UMR1037-INSERM, ERL5294 CNRS, Toulouse, France.,Laboratoire d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France
| | - Thibault Comont
- Service de Médecine Interne, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France
| | - Audrey Sarry
- Service d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France
| | - Françoise Huguet
- Service d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France
| | - Emilie Bérard
- Service d'Epidémiologie, Centre Hospitalier Universitaire de Toulouse, Toulouse, France.,UMR 1027, INSERM-Université de Toulouse III, Toulouse, France
| | - Christian Récher
- Service d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France.,Université Toulouse III Paul Sabatier, Toulouse, France.,Cancer Research Center of Toulouse, UMR1037-INSERM, ERL5294 CNRS, Toulouse, France
| |
Collapse
|