1
|
Archer KJ, Fu H, Mrózek K, Nicolet D, Mims AS, Uy GL, Stock W, Byrd JC, Hiddemann W, Braess J, Spiekermann K, Metzeler KH, Herold T, Eisfeld AK. Identifying long-term survivors and those at higher or lower risk of relapse among patients with cytogenetically normal acute myeloid leukemia using a high-dimensional mixture cure model. J Hematol Oncol 2024; 17:28. [PMID: 38702786 PMCID: PMC11068580 DOI: 10.1186/s13045-024-01553-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/29/2024] [Indexed: 05/06/2024] Open
Abstract
Patients with cytogenetically normal acute myeloid leukemia (CN-AML) may harbor prognostically relevant gene mutations and thus be categorized into one of the three 2022 European LeukemiaNet (ELN) genetic-risk groups. Nevertheless, there remains heterogeneity with respect to relapse-free survival (RFS) within these genetic-risk groups. Our training set included 306 adults on Alliance for Clinical Trials in Oncology studies with de novo CN-AML aged < 60 years who achieved a complete remission and for whom centrally reviewed cytogenetics, RNA-sequencing, and gene mutation data from diagnostic samples were available (Alliance trial A152010). To overcome deficiencies of the Cox proportional hazards model when long-term survivors are present, we developed a penalized semi-parametric mixture cure model (MCM) to predict RFS where RNA-sequencing data comprised the predictor space. To validate model performance, we employed an independent test set from the German Acute Myeloid Leukemia Cooperative Group (AMLCG) consisting of 40 de novo CN-AML patients aged < 60 years who achieved a complete remission and had RNA-sequencing of their pre-treatment sample. For the training set, there was a significant non-zero cure fraction (p = 0.019) with 28.5% of patients estimated to be cured. Our MCM included 112 genes associated with cure, or long-term RFS, and 87 genes associated with latency, or shorter-term time-to-relapse. The area under the curve and C-statistic were respectively, 0.947 and 0.783 for our training set and 0.837 and 0.718 for our test set. We identified a novel, prognostically relevant molecular signature in CN-AML, which allows identification of patient subgroups independent of 2022 ELN genetic-risk groups.Trial registration Data from companion studies CALGB 8461, 9665 and 20202 (trials registered at www.clinicaltrials.gov as, respectively, NCT00048958, NCT00899223, and NCT00900224) were obtained from Alliance for Clinical Trials in Oncology under data sharing study A152010. Data from the AMLCG 2008 trial was registered at www.clinicaltrials.gov as NCT01382147.
Collapse
Affiliation(s)
- Kellie J Archer
- Division of Biostatistics, College of Public Health, The Ohio State University, 240 Cunz Hall, 1841 Neil Avenue, Columbus, OH, 43210, USA.
| | - Han Fu
- Google, Inc., Mountain View, CA, USA
| | - Krzysztof Mrózek
- Clara D. Bloomfield Center for Leukemia Outcomes Research, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Deedra Nicolet
- Clara D. Bloomfield Center for Leukemia Outcomes Research, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
- Alliance Statistics and Data Management Center, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Alice S Mims
- Clara D. Bloomfield Center for Leukemia Outcomes Research, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Geoffrey L Uy
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Wendy Stock
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - John C Byrd
- Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Wolfgang Hiddemann
- Laboratory for Leukemia Diagnostics, Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Jan Braess
- Department of Oncology and Hematology, Hospital Barmherzige Brüder, Regensburg, Germany
| | - Karsten Spiekermann
- Laboratory for Leukemia Diagnostics, Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Klaus H Metzeler
- Department of Hematology and Cellular Therapy, University Hospital Leipzig, Leipzig, Germany
| | - Tobias Herold
- Laboratory for Leukemia Diagnostics, Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Ann-Kathrin Eisfeld
- Clara D. Bloomfield Center for Leukemia Outcomes Research, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| |
Collapse
|
2
|
Schlenk RF, Weber D, Krzykalla J, Kindler T, Wulf G, Hertenstein B, Salih HR, Südhoff T, Krauter J, Martens U, Wessendorf S, Runde V, Tischler HJ, Bentz M, Koller E, Heuser M, Thol F, Benner A, Ganser A, Döhner K, Döhner H. Randomized phase-III study of low-dose cytarabine and etoposide + /- all-trans retinoic acid in older unfit patients with NPM1-mutated acute myeloid leukemia. Sci Rep 2023; 13:14809. [PMID: 37684299 PMCID: PMC10491626 DOI: 10.1038/s41598-023-41964-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 09/04/2023] [Indexed: 09/10/2023] Open
Abstract
The aim of this randomized clinical trial was to evaluate the impact of all-trans retinoic acid (ATRA) in combination with non-intensive chemotherapy in older unfit patients (> 60 years) with newly diagnosed NPM1-mutated acute myeloid leukemia. Patients were randomized (1:1) to low-dose chemotherapy with or without open-label ATRA 45 mg/m2, days 8-28; the dose of ATRA was reduced to 45 mg/m2, days 8-10 and 15 mg/m2, days 11-28 after 75 patients due to toxicity. Up to 6 cycles of cytarabine 20 mg/day s.c., bid, days 1-7 and etoposide 100 mg/day, p.o. or i.v., days 1-3 with (ATRA) or without ATRA (CONTROL) were intended. The primary endpoint was overall survival (OS). Between May 2011 and September 2016, 144 patients (median age, 77 years; range, 64-92 years) were randomized (72, CONTROL; 72, ATRA). Baseline characteristics were balanced between the two study arms. The median number of treatment cycles was 2 in ATRA and 2.5 in CONTROL. OS was significantly shorter in the ATRA compared to the CONTROL arm (p = 0.023; median OS: 5 months versus 9.2 months, 2-years OS rate: 7% versus 10%, respectively). Rates of CR/CRi were not different between treatment arms; infections were more common in ATRA beyond treatment cycle one. The addition of ATRA to low-dose cytarabine plus etoposide in an older, unfit patient population was not beneficial, but rather led to an inferior outcome.The clinical trial is registered at clinicaltrialsregister.eu (EudraCT Number: 2010-023409-37, first posted 14/12/2010).
Collapse
Affiliation(s)
- R F Schlenk
- NCT-Trial Center, National Center of Tumor Diseases, Heidelberg University Hospital and German Cancer Research Center, Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany.
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.
| | - D Weber
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - J Krzykalla
- Division of Biostatistics, German Cancer Research Center Heidelberg, Heidelberg, Germany
| | - T Kindler
- Department of Hematology, Medical Oncology and Pneumology, University Medical Center Mainz, Mainz, Germany
| | - G Wulf
- Department of Hematology and Oncology, University Hospital of Göttingen, Göttingen, Germany
| | - B Hertenstein
- Department of Hematology and Oncology, Klinikum Bremen Mitte, Bremen, Germany
| | - H R Salih
- Department of Hematology and Oncology, Eberhard-Karls University, Tübingen, Germany
| | - T Südhoff
- Department of Hematology and Oncology, Klinikum Passau, Passau, Germany
| | - J Krauter
- Department Hematology and Oncology, Braunschweig Municipal Hospital, Braunschweig, Germany
| | - U Martens
- Department of Hematology and Oncology, Klinikum am Gesundbrunnen, Heilbronn, Germany
| | - S Wessendorf
- Department of Hematology and Oncology, Klinikum Esslingen, Esslingen, Germany
| | - V Runde
- Department of Hematology/Oncology, Wilhelm-Anton Hospital Goch, Goch, Germany
| | - H J Tischler
- Department of Hematology and Oncology, University Hospital of Minden, Minden, Germany
| | - M Bentz
- Department of Hematology and Oncology, Städtisches Klinikum Karlsruhe, Karlsruhe, Germany
| | - E Koller
- Department of Internal Medicine III, Hanuschkrankenhaus Wien, Wien, Austria
| | - M Heuser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - F Thol
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - A Benner
- Division of Biostatistics, German Cancer Research Center Heidelberg, Heidelberg, Germany
| | - A Ganser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - K Döhner
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - H Döhner
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| |
Collapse
|
3
|
Jiao W, Liu Y, Bao Y. Clinical Significance of EZH2 in Acute Myeloid Leukemia. COMPUTATIONAL INTELLIGENCE AND NEUROSCIENCE 2022; 2022:8741989. [PMID: 36052036 PMCID: PMC9427216 DOI: 10.1155/2022/8741989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 11/17/2022]
Abstract
In this paper, we have focused on the investigation of the expression level of the enhancer of zeste homolog 2 (EZH2) gene in bone marrow mononuclear cells of acute myeloid leukemia (AML) patients and analyze the relationship between EZH2 gene expression and EMI. The expression of EZH2mRNA in bone marrow mononuclear cells of 26 patients with incipient AML was detected by qRT-PCR, and the relationship between EZH2mRNA expression and clinical characteristics was analyzed. EZH2 mRNA expression was increased in 26 AML patients. EZH2 gene expression in male patients was significantly higher than that in female patients. The expression of EZH2 in the group with extramedullary infiltration (EMI) was significantly higher than that in the group without EMI. The patients were divided into different groups according to the chromosomal karyotype and prognosis. Statistical analysis showed that the expression level of the medium-risk group was significantly higher than that of the low-risk group, while there was no statistical difference in other groups (P > 0.05). The expression of EZH2 gene in AML patients was closely related to EMI, and the expression of EZH2 in AML cells was closely related to cell migration ability. EZH2 is expected to be one of the indicators of disease recurrence.
Collapse
Affiliation(s)
- Weiyun Jiao
- Hematology Department, Hefei First People's Hospital, Hefei 23000, China
| | - Yuanyuan Liu
- Hematology Department, Hefei First People's Hospital, Hefei 23000, China
| | - Yangyi Bao
- Hematology Department, Hefei First People's Hospital, Hefei 23000, China
| |
Collapse
|
4
|
Yılmaz H, Toy HI, Marquardt S, Karakülah G, Küçük C, Kontou PI, Logotheti S, Pavlopoulou A. In Silico Methods for the Identification of Diagnostic and Favorable Prognostic Markers in Acute Myeloid Leukemia. Int J Mol Sci 2021; 22:ijms22179601. [PMID: 34502522 PMCID: PMC8431757 DOI: 10.3390/ijms22179601] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/13/2021] [Accepted: 08/20/2021] [Indexed: 12/13/2022] Open
Abstract
Acute myeloid leukemia (AML), the most common type of acute leukemia in adults, is mainly asymptomatic at early stages and progresses/recurs rapidly and frequently. These attributes necessitate the identification of biomarkers for timely diagnosis and accurate prognosis. In this study, differential gene expression analysis was performed on large-scale transcriptomics data of AML patients versus corresponding normal tissue. Weighted gene co-expression network analysis was conducted to construct networks of co-expressed genes, and detect gene modules. Finally, hub genes were identified from selected modules by applying network-based methods. This robust and integrative bioinformatics approach revealed a set of twenty-four genes, mainly related to cell cycle and immune response, the diagnostic significance of which was subsequently compared against two independent gene expression datasets. Furthermore, based on a recent notion suggesting that molecular characteristics of a few, unusual patients with exceptionally favorable survival can provide insights for improving the outcome of individuals with more typical disease trajectories, we defined groups of long-term survivors in AML patient cohorts and compared their transcriptomes versus the general population to infer favorable prognostic signatures. These findings could have potential applications in the clinical setting, in particular, in diagnosis and prognosis of AML.
Collapse
Affiliation(s)
- Hande Yılmaz
- Izmir Biomedicine and Genome Center, Balcova, 35340 Izmir, Turkey; (H.Y.); (H.I.T.); (G.K.); (C.K.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Balcova, 35340 Izmir, Turkey
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, 18057 Rostock, Germany;
| | - Halil Ibrahim Toy
- Izmir Biomedicine and Genome Center, Balcova, 35340 Izmir, Turkey; (H.Y.); (H.I.T.); (G.K.); (C.K.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Balcova, 35340 Izmir, Turkey
| | - Stephan Marquardt
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, 18057 Rostock, Germany;
| | - Gökhan Karakülah
- Izmir Biomedicine and Genome Center, Balcova, 35340 Izmir, Turkey; (H.Y.); (H.I.T.); (G.K.); (C.K.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Balcova, 35340 Izmir, Turkey
| | - Can Küçük
- Izmir Biomedicine and Genome Center, Balcova, 35340 Izmir, Turkey; (H.Y.); (H.I.T.); (G.K.); (C.K.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Balcova, 35340 Izmir, Turkey
- Department of Medical Biology, Faculty of Medicine, Dokuz Eylül University, Balcova, 35340 Izmir, Turkey
| | - Panagiota I. Kontou
- Department of Computer Science and Biomedical Informatics, University of Thessaly, 35131 Lamia, Greece;
| | - Stella Logotheti
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, 18057 Rostock, Germany;
- Correspondence: (S.L.); (A.P.)
| | - Athanasia Pavlopoulou
- Izmir Biomedicine and Genome Center, Balcova, 35340 Izmir, Turkey; (H.Y.); (H.I.T.); (G.K.); (C.K.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Balcova, 35340 Izmir, Turkey
- Correspondence: (S.L.); (A.P.)
| |
Collapse
|
5
|
Derissen EJB, Beijnen JH. Intracellular Pharmacokinetics of Pyrimidine Analogues used in Oncology and the Correlation with Drug Action. Clin Pharmacokinet 2020; 59:1521-1550. [PMID: 33064276 PMCID: PMC7717039 DOI: 10.1007/s40262-020-00934-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pyrimidine analogues can be considered as prodrugs, like their natural counterparts, they have to be activated within the cell. The intracellular activation involves several metabolic steps including sequential phosphorylation to its monophosphate, diphosphate and triphosphate. The intracellularly formed nucleotides are responsible for the pharmacological effects. This review provides a comprehensive overview of the clinical studies that measured the intracellular nucleotide concentrations of pyrimidine analogues in patients with cancer. The objective was to gain more insight into the parallels between the different pyrimidine analogues considering their intracellular pharmacokinetics. For cytarabine and gemcitabine, the intracellular pharmacokinetics have been extensively studied over the years. However, for 5-fluorouracil, capecitabine, azacitidine and decitabine, the intracellular pharmacokinetics was only very minimally investigated. This is probably owing to the fact that there were no suitable bioanalytical assays for a long time. Since the advent of suitable assays, the first exploratory studies indicate that the intracellular 5-fluorouracil, azacitidine and decitabine nucleotide concentrations are very low compared with the intracellular nucleotide concentrations obtained during treatment with cytarabine or gemcitabine. Based on their pharmacology, the intracellular accumulation of nucleotides appears critical to the cytotoxicity of pyrimidine analogues. However, not many clinical studies have actually investigated the relationship between the intracellular nucleotide concentrations in patients with cancer and the anti-tumour effect. Only for cytarabine, a relationship was demonstrated between the intracellular triphosphate concentrations in leukaemic cells and the response rate in patients with AML. Future clinical studies should show, for the other pyrimidine analogues, whether there is a relationship between the intracellular nucleotide concentrations and the clinical outcome of patients. Research that examined the intracellular pharmacokinetics of cytarabine and gemcitabine focused primarily on the saturation aspect of the intracellular triphosphate formation. Attempts to improve the dosing regimen of gemcitabine were aimed at maximising the intracellular gemcitabine triphosphate concentrations. However, this strategy does not make sense, as efficient administration also means that less gemcitabine can be administered before dose-limiting toxicities are achieved. For all pyrimidine analogues, a linear relationship was found between the dose and the plasma concentration. However, no correlation was found between the plasma concentration and the intracellular nucleotide concentration. The concentration-time curves for the intracellular nucleotides showed considerable inter-individual variation. Therefore, the question arises whether pyrimidine analogue therapy should be more individualised. Future research should show which intracellular nucleotide concentrations are worth pursuing and whether dose individualisation is useful to achieve these concentrations.
Collapse
Affiliation(s)
- Ellen J B Derissen
- Department of Pharmacy and Pharmacology, Antoni van Leeuwenhoek Hospital-The Netherlands Cancer Institute, Louwesweg 6, 1066 EC , Amsterdam, The Netherlands. .,Department of Clinical Pharmacology and Pharmacy, Amsterdam UMC, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands. .,Department of Pharmacy , Elisabeth-TweeSteden Hospital, Dr. Deelenlaan 5, 5042 AD, Tilburg, The Netherlands.
| | - Jos H Beijnen
- Department of Pharmacy and Pharmacology, Antoni van Leeuwenhoek Hospital-The Netherlands Cancer Institute, Louwesweg 6, 1066 EC , Amsterdam, The Netherlands.,Science Faculty, Division of Pharmaco-epidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, P.O. Box 80082, 3508 TB, Utrecht, The Netherlands
| |
Collapse
|
6
|
Borlenghi E, Cattaneo C, Cerqui E, Archetti S, Bertoli D, Bellotti D, Gramegna D, Soverini G, Oberti M, Schieppati F, Pagani C, Passi A, Sciumé M, Farina M, Carbone C, Crippa C, Dalceggio D, Tucci A, Rossi G. Postremission therapy with repeated courses of high-dose cytarabine, idarubicin, and limited autologous stem cell support achieves a very good long-term outcome in European leukemia net favorable and intermediate-risk acute myeloid leukemia. Hematol Oncol 2020; 38:754-762. [PMID: 32950042 DOI: 10.1002/hon.2806] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/16/2020] [Accepted: 09/18/2020] [Indexed: 12/17/2022]
Abstract
Consolidation treatment in acute myeloid leukemia (AML) patients achieving complete remission (CR) is warranted. High-dose cytarabine (HDAC) is considered first choice in favorable risk and an option in intermediate-risk AML. However, its optimal dose and schedule, as well as the benefit of additional chemotherapy agents remain controversial. Herein, we report on the long-term outcome of consecutive unselected AML patients treated with repeated courses of HDAC, with the addition of idarubicin, followed by autologous peripheral blood stem cell (PBSC) support, in order to limit toxicity, according to Northern Italy Leukemia Group (NILG) AML-01/00 study (EUDRACT number 00400673). Among 338 patients consecutively diagnosed from 2001 to 2017 at our center, 148 with high-risk AML (adverse cytogenetic, isolated FLT3-internal tandem duplication mutation, refractory to first induction) were addressed to allogeneic stem cell transplant. All other cases, 186 patients (55%), median age 53 (range 19-75), were considered standard-risk and received the NILG AML-01/00 program. After achieving CR, patients were mobilized with cytarabine 8 g/sqm to collect autologous CD34+-PBSC and received three consolidation cycles with HDAC (20 g/sqm) plus idarubicin (20 mg/sqm) per cycle, followed by reinfusion of limited doses of CD34+ PBSC (1-2x106/kg). The program was completed by 160 (86%) patients. Toxicity was acceptable. Neutrophils recovered a median of 10 days. Treatment-related mortality was 3/160 (1.8%). After a median follow-up of 66.4 months, overall survival (OS) and relapse-free survival (RFS) at 5-years were 61.4% and 52.4%, respectively. Twenty-eight selected patients aged >65 had similar outcomes. According to European leukemia net-2010 classification, the OS and RFS at 5-years were 76.4% and 65% in favorable risk, without differences between molecular subgroups, 52.3% and 47.2% in Intermediate-I, 45.2% and 36.5% in Intermediate-II risk patients, respectively. In conclusion, consolidation including repeated courses of high dose cytarabine and idarubicin, with limited PBSC support, proved feasible and very effective in nonhigh risk patients. The incorporation of novel agents in its backbone may be tested to further improve patient's prognosis.
Collapse
Affiliation(s)
| | | | - Elisa Cerqui
- Hematology Department, ASST Spedali Civili, Brescia, Italy
| | - Silvana Archetti
- Diagnostic Department, Clinical Chemistry Laboratory, ASST Spedali Civili, Brescia, Italy
| | - Diego Bertoli
- Diagnostic Department, Clinical Chemistry Laboratory, ASST Spedali Civili, Brescia, Italy
| | - Daniela Bellotti
- Laboratorio di Citogenetica e Genetica Medica, ASST Spedali Civili, Brescia, Italy
| | | | | | | | - Francesca Schieppati
- Immunohematology and Trasfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
| | - Chiara Pagani
- Hematology Department, ASST Spedali Civili, Brescia, Italy
| | - Angela Passi
- Hematology Department, ASST Spedali Civili, Brescia, Italy
| | | | - Mirko Farina
- Hematology Department, ASST Spedali Civili, Brescia, Italy
| | | | - Claudia Crippa
- Hematology Department, ASST Spedali Civili, Brescia, Italy
| | | | | | - Giuseppe Rossi
- Hematology Department, ASST Spedali Civili, Brescia, Italy
| |
Collapse
|
7
|
Tallman MS, Wang ES, Altman JK, Appelbaum FR, Bhatt VR, Bixby D, Coutre SE, De Lima M, Fathi AT, Fiorella M, Foran JM, Hall AC, Jacoby M, Lancet J, LeBlanc TW, Mannis G, Marcucci G, Martin MG, Mims A, O'Donnell MR, Olin R, Peker D, Perl A, Pollyea DA, Pratz K, Prebet T, Ravandi F, Shami PJ, Stone RM, Strickland SA, Wieduwilt M, Gregory KM, Hammond L, Ogba N. Acute Myeloid Leukemia, Version 3.2019, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2020; 17:721-749. [PMID: 31200351 DOI: 10.6004/jnccn.2019.0028] [Citation(s) in RCA: 308] [Impact Index Per Article: 61.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Acute myeloid leukemia (AML) is the most common form of acute leukemia among adults and accounts for the largest number of annual deaths due to leukemias in the United States. Recent advances have resulted in an expansion of treatment options for AML, especially concerning targeted therapies and low-intensity regimens. This portion of the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for AML focuses on the management of AML and provides recommendations on the workup, diagnostic evaluation and treatment options for younger (age <60 years) and older (age ≥60 years) adult patients.
Collapse
Affiliation(s)
| | | | - Jessica K Altman
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | | | | | - Dale Bixby
- University of Michigan Rogel Cancer Center
| | | | - Marcos De Lima
- Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | | | | | | | - Aric C Hall
- University of Wisconsin Carbone Cancer Center
| | - Meagan Jacoby
- Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | | | | | | | | | - Michael G Martin
- St. Jude Children's Research Hospital/The University of Tennessee Health Science Center
| | - Alice Mims
- The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | | | - Rebecca Olin
- UCSF Helen Diller Family Comprehensive Cancer Center
| | | | - Alexander Perl
- Abramson Cancer Center at the University of Pennsylvania
| | | | - Keith Pratz
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | | | | | - Paul J Shami
- Huntsman Cancer Institute at the University of Utah
| | | | | | | | | | -
- National Comprehensive Cancer Network
| | | | | |
Collapse
|
8
|
Nagahata Y, Kondo T, Ono Y, Hiramoto N, Kitano T, Hishizawa M, Yamashita K, Hashimoto H, Ishikawa T, Takaori-Kondo A. High-dose cytarabine chemotherapy (≥4 g/m 2/day) before allogeneic hematopoietic stem cell transplantation for non-core-binding-factor AML in the first complete remission. Leuk Lymphoma 2020; 61:3128-3136. [PMID: 32804016 DOI: 10.1080/10428194.2020.1805112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Benefit of high-dose cytarabine (HD-AraC) for acute myeloid leukemia (AML) prior to allogeneic hematopoietic stem cell transplantation (allo-HSCT) remains unknown. We retrospectively analyzed data from 79 non-core-binding-factor AML patients who underwent allo-HSCT in their first complete remission (CR1). In univariate analysis, HD-AraC (≥4 g/m2/day) before allo-HSCT improved disease-free survival (DFS) (p = .018), overall survival (OS) (p = .029), and cumulative incidence of relapse (CIR) (p = .033). Four-year DFS, OS, and CIR of patients receiving and not receiving HD-AraC were 79% vs. 49%, 82% vs. 56%, and 18% vs. 42%, respectively. In multivariate analysis, HD-AraC was a positive prognostic factor for DFS (hazard ratio (HR) = 0.36, 95% confidence interval (CI): 0.14-0.88), OS (HR = 0.37, 95% CI: 0.14-0.99), and CIR (HR = 0.38, 95% CI; 0.14-1.0). Our study demonstrates that HD-AraC before allo-HSCT at a dose ≥4 g/m2/day is effective for treating AML patients in CR1.
Collapse
Affiliation(s)
- Yosuke Nagahata
- Department of Hematology/Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tadakazu Kondo
- Department of Hematology/Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yuichiro Ono
- Department of Hematology, Kobe Medical Center General Hospital, Kobe, Japan
| | - Nobuhiro Hiramoto
- Department of Cell Therapy, Institute of Biomedical Research and Innovation, Kobe, Japan
| | - Toshiyuki Kitano
- Department of Hematology/Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masakatsu Hishizawa
- Department of Hematology/Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kouhei Yamashita
- Department of Hematology/Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hisako Hashimoto
- Department of Cell Therapy, Institute of Biomedical Research and Innovation, Kobe, Japan
| | - Takayuki Ishikawa
- Department of Hematology, Kobe Medical Center General Hospital, Kobe, Japan
| | - Akifumi Takaori-Kondo
- Department of Hematology/Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
9
|
Next Generation Sequencing in AML-On the Way to Becoming a New Standard for Treatment Initiation and/or Modulation? Cancers (Basel) 2019; 11:cancers11020252. [PMID: 30795628 PMCID: PMC6406956 DOI: 10.3390/cancers11020252] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/08/2019] [Accepted: 02/12/2019] [Indexed: 12/19/2022] Open
Abstract
Acute myeloid leukemia (AML) is a clonal disease caused by genetic abberations occurring predominantly in the elderly. Next generation sequencing (NGS) analysis has led to a deeper genetic understanding of the pathogenesis and the role of recently discovered genetic precursor lesions (clonal hematopoiesis of indeterminate/oncogenic potential (CHIP/CHOP)) in the evolution of AML. These advances are reflected by the inclusion of certain mutations in the updated World Health Organization (WHO) 2016 classification and current treatment guidelines by the European Leukemia Net (ELN) and National Comprehensive Cancer Network (NCCN) and results of mutational testing are already influencing the choice and timing of (targeted) treatment. Genetic profiling and stratification of patients into molecularly defined subgroups are expected to gain ever more weight in daily clinical practice. Our aim is to provide a concise summary of current evidence regarding the relevance of NGS for the diagnosis, risk stratification, treatment planning and response assessment in AML, including minimal residual disease (MRD) guided approaches. We also summarize recently approved drugs targeting genetically defined patient populations with risk adapted- and individualized treatment strategies.
Collapse
|
10
|
Mueller BU, Seipel K, Pabst T. Myelodysplastic syndromes and acute myeloid leukemias in the elderly. Eur J Intern Med 2018; 58:28-32. [PMID: 30527920 DOI: 10.1016/j.ejim.2018.05.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 05/20/2018] [Accepted: 05/23/2018] [Indexed: 01/29/2023]
Abstract
Most patients above 60 years with acute myeloid leukemia (AML) will die from their disease. Nevertheless, the treatment concepts in elderly patients with myelodysplastic syndromes (MDS) and AML are rapidly evolving. A number of recent reports have identified better survival rates with intensive induction chemotherapy for patients up to 80 years, with the exception of patients with unfavorable genomic risk abnormalities or with major co-morbidities. Gemtuzumab ozogamicin is increasingly added to induction therapy for AML patients up to 70 years with favorable or intermediate risk profile, and Midostaurin for patients with a FLT3 mutation. The recommended dose of daunorubicin is 60 mg/m2 for 3 + 7 induction therapy. Elderly patients with acute promyelocytic leukemia should receive all-trans retinoic acid and arsenic trioxide, and cytotoxic treatment is limited upfront to patients with initial leukocytosis. Allogeneic transplantation can be recommended to selected patients up to 70-75 years. For patients unfit for intensive treatment, therapeutic options comprise a hypomethylating agent (HMA), low-dose cytarabin and supportive care. HMA treatment is also increasingly applied for relapsed/refractory AML after intensive chemotherapy. A considerable number of candidate compounds are currently being studied in older AML patients, with their potential role in the treatment of elderly AML patients remaining to be clarified.
Collapse
Affiliation(s)
- Beatrice U Mueller
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Katja Seipel
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Thomas Pabst
- Department of Medical Oncology, Inselspital, University Hospital and University of Bern, Bern, Switzerland.
| |
Collapse
|
11
|
Visani G, Loscocco F, Isidori A, Piccaluga PP. Genetic profiling in acute myeloid leukemia: a path to predicting treatment outcome. Expert Rev Hematol 2018; 11:455-461. [PMID: 29792762 DOI: 10.1080/17474086.2018.1475225] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 05/08/2018] [Indexed: 10/16/2022]
Abstract
Despite substantial progresses in acute myeloid leukemia (AML) diagnosis and treatment, at least half of patient will eventually die for the disease. In the last decades, the use of genetic and genomic approaches allowed the identification of patients with higher risk of recurrence after and/or resistance to CHT. However, though many novel drugs have been proposed and tested, only little clinical improvements have been made concerning the treatment of the so called 'high risk' patients. Areas covered: In this article, the authors, based on their own experience and the most updated literature, review the basic knowledge of AML prognostication and treatment prediction developed throughout genetic and genomic profiling, and focus on the use of gene expression profiling as a promising predictive tool. The role of next generation sequencing, run on qPCR/digital PCR platforms or polyvalent ones such as the Nanostring NCounter™ and RNA-sequencing techniques in the near future will also be briefly discussed. Expert commentary: The authors believe that a combination of genetic (including both germline and somatic data), epigenetic and transcriptional data will represent, in the future, the molecular basis for treatment decision with the highest predictive potential.
Collapse
MESH Headings
- DNA Mutational Analysis/methods
- Drug Resistance, Neoplasm
- Epigenesis, Genetic
- Gene Expression Regulation, Leukemic
- Genetic Profile
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Transcription, Genetic
Collapse
Affiliation(s)
- Giuseppe Visani
- a Hematology and Hematopoietic Stem Cell Transplant Center , AORMN , Pesaro , Italy
| | - Federica Loscocco
- a Hematology and Hematopoietic Stem Cell Transplant Center , AORMN , Pesaro , Italy
| | - Alessandro Isidori
- a Hematology and Hematopoietic Stem Cell Transplant Center , AORMN , Pesaro , Italy
| | - Pier Paolo Piccaluga
- b Department of Experimental, Diagnostic, and Specialty Medicine, S. Orsola-Malpighi Hospital , Bologna University School of Medicine , Bologna , Italy
- c Euro-Mediterranean Institute of Science and Technology (IEMEST) , Palermo , Italy
- d Department of Pathology , Jomo Kenyatta University of Agriculture and Technology , Nairobi , Kenya
| |
Collapse
|
12
|
Li XX, Guo H, Zhou JD, Wu DH, Ma JC, Wen XM, Zhang W, Xu ZJ, Lin J, Jun Q. Overexpression of CTNNB1: Clinical implication in Chinese de novo acute myeloid leukemia. Pathol Res Pract 2018; 214:361-367. [PMID: 29496308 DOI: 10.1016/j.prp.2018.01.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 12/25/2017] [Accepted: 01/05/2018] [Indexed: 12/28/2022]
Abstract
Activation of Wnt/β-catenin signaling played a crucial role in tumorigenesis, and β-catenin (CTNNB1) overexpression has been identified in numerous solid tumors. The present study was designed to determine CTNNB1 expression and its clinical significance in Chinese de novo acute myeloid leukemia (AML) patients. Real-time quantitative PCR was carried out to detect the pattern of CTNNB1 expression in 140 AML patients and 46 controls. The level of CTNNB1 transcript in AML patients was significantly up-regulated compared with controls (P < 0.001). CTNNB1high patients showed significantly older age than CTNNB1low patients (P < 0.05). The frequency of high CTNNB1 expression was significantly observed in patients with intermediate/poor karyotypes. CTNNB1high patients had a significantly lower complete remission (CR) rate than CTNNB1low patients (P = 0.004). Among cytogenetically normal AML (CN-AML), CTNNB1high patients presented significantly shorter overall survival (OS, P = 0.004) and leukemia-free survival (LFS, P = 0.038) than CTNNB1low patients. Multivariate analysis confirmed that CTNNB1 expression was an independent prognostic factor for OS among CN-AML. Moreover, CTNNB1 expression level significantly decreased after CR stage (P = 0.032) and increased in relapsed stage (P = 0.015). Our findings suggest that CTNNB1 is overexpressed and confers a poor prognosis in AML, and could be used as a biomarker in monitoring disease recurrence.
Collapse
Affiliation(s)
- Xi-Xi Li
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China; The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China; School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Hong Guo
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China; The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China
| | - Jing-Dong Zhou
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China; The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China; School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - De-Hong Wu
- Department of Hematology, The Third People's Hospital of KunShan City, Suzhou, Jiangsu, People's Republic of China
| | - Ji-Chun Ma
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China; The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China
| | - Xiang-Mei Wen
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China; The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China
| | - Wei Zhang
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China; The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China
| | - Zi-Jun Xu
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China; The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China; School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Jiang Lin
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China; The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China
| | - Qian Jun
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China; The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China.
| |
Collapse
|
13
|
Luskin MR, DeAngelo DJ. Midostaurin/PKC412 for the treatment of newly diagnosed FLT3 mutation-positive acute myeloid leukemia. Expert Rev Hematol 2017; 10:1033-1045. [PMID: 29069942 DOI: 10.1080/17474086.2017.1397510] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Acute myeloid leukemia (AML) is an aggressive hematologic malignancy with inadequate treatment options. Approximately one-third of cases have a FLT3-ITD or FLT3-TKD mutation which leads to constitutive tyrosine kinase activation which contributes to leukemogenesis. The FLT3-ITD mutation is associated with a particularly poor prognosis. Midostaurin is a multi-kinase inhibitor active against the FLT3 receptor. Midostaurin was approved by the US FDA in April 2017 for treatment of newly diagnosed FLT3-mutant AML in combination with chemotherapy. Areas covered: Standard treatment of FLT3-mutant AML and outcomes. Early clinical development of midostaurin including pharmacokinetics and metabolism. The development of midostaurin in FLT3-mutant AML is then outlined including review of the phase I, II, and III trials of midostaurin as a single agent and in combination with chemotherapy. Expert commentary: The approval of midostaurin represents the first new therapy for AML in several decades. It is also the first targeted therapy approved for AML. Future studies will focus on defining mechanisms of resistance to midostaurin as well as establishing the role of midostaurin in combination with hypomethylating agents and as maintenance therapy. Second generation, more potent and selective FLT3 inhibitors are also in development; these agents need to be compared to midostaurin.
Collapse
Affiliation(s)
- Marlise R Luskin
- a Department of Medical Oncology , Dana-Farber Cancer Institute , Boston , MA , USA.,b Harvard Medical School , Boston , MA , USA
| | - Daniel J DeAngelo
- a Department of Medical Oncology , Dana-Farber Cancer Institute , Boston , MA , USA.,b Harvard Medical School , Boston , MA , USA
| |
Collapse
|
14
|
Small molecule inhibitors for acute myeloid leukemia: where is the field heading? Future Med Chem 2017; 9:1453-1456. [DOI: 10.4155/fmc-2017-0114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
|
15
|
Damlaj M, Ghazi S, Mashaqbeh W, Gmati G, Salama H, Abuelgasim KA, Rather M, Hajeer A, Al-Zahrani M, Jazieh AR, Hejazi A, Al Askar A. Lymphocyte recovery is an independent predictor of relapse in allogeneic hematopoietic cell transplantation recipients for acute leukemia. World J Transplant 2017; 7:235-242. [PMID: 28900606 PMCID: PMC5573899 DOI: 10.5500/wjt.v7.i4.235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 07/06/2017] [Accepted: 07/24/2017] [Indexed: 02/05/2023] Open
Abstract
AIM To examine the optimal absolute lymphocyte count (ALC) cut-off utilizing receiver operator characteristics (ROC) in addition to graft characteristics associated with early ALC recovery.
METHODS Patients who received T-cell replete peripheral hematopoietic cell transplantation (HCT) for acute leukemia were identified. ALC cut-off was established using ROC analysis and subsequently the cohort was stratified. Time to endpoint analysis and cox regression modelling was computed to analyze outcomes.
RESULTS A total of 72 patients met the inclusion criteria and were analyzed. Optimal ALC cut-off was established to be on day 14 (D14) with ALC > 0.3 × 109/L. At 2 years, cumulative incidence of relapse was 16.9% vs 46.9% (P = 0.025) for early and delayed lymphocyte recovery cohorts, respectively. Chronic graft vs host disease was more prevalent in the early lymphocyte recovery (ELR) group at 70% vs 27%, respectively (P = 0.0006). On multivariable analysis for relapse, ELR retained its prognostic significance with HR = 0.27 (0.05-0.94, P = 0.038).
CONCLUSION ELR is an independent predictor for relapse in patients receiving allogeneic HCT for acute leukemia. ELR was influenced by graft characteristics particularly CD34 count.
Collapse
Affiliation(s)
- Moussab Damlaj
- Division of Hematology and HSCT, Department of Oncology, King Abdulaziz Medical City, Riyadh 11426, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia
| | - Samer Ghazi
- Division of Hematology and HSCT, Department of Oncology, King Abdulaziz Medical City, Riyadh 11426, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia
| | - Walid Mashaqbeh
- Division of Hematology and HSCT, Department of Oncology, King Abdulaziz Medical City, Riyadh 11426, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia
| | - Gamal Gmati
- Division of Hematology and HSCT, Department of Oncology, King Abdulaziz Medical City, Riyadh 11426, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia
| | - Hend Salama
- Division of Hematology and HSCT, Department of Oncology, King Abdulaziz Medical City, Riyadh 11426, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia
| | - Khadega A Abuelgasim
- Division of Hematology and HSCT, Department of Oncology, King Abdulaziz Medical City, Riyadh 11426, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia
| | - Mushtaq Rather
- Division of Hematology and HSCT, Department of Oncology, King Abdulaziz Medical City, Riyadh 11426, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia
| | - Ali Hajeer
- King Saud bin Abdulaziz University for Health Sciences, Riyadh 11426, Saudi Arabia
| | - Mohsen Al-Zahrani
- Division of Hematology and HSCT, Department of Oncology, King Abdulaziz Medical City, Riyadh 11426, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia
| | - Abdul-Rahman Jazieh
- Division of Hematology and HSCT, Department of Oncology, King Abdulaziz Medical City, Riyadh 11426, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia
| | - Ayman Hejazi
- Division of Hematology and HSCT, Department of Oncology, King Abdulaziz Medical City, Riyadh 11426, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia
| | - Ahmad Al Askar
- Division of Hematology and HSCT, Department of Oncology, King Abdulaziz Medical City, Riyadh 11426, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia
| |
Collapse
|
16
|
Benicio MTDL, Ribeiro AFT, Américo AD, Furtado FM, Glória AB, Lima AS, Santos SM, Xavier SG, Lucena-Araujo AR, Fagundes EM, Rego EM. Evaluation of the European LeukemiaNet recommendations for predicting outcomes of patients with acute myeloid leukemia treated in low- and middle-income countries (LMIC): A Brazilian experience. Leuk Res 2017; 60:109-114. [PMID: 28777950 DOI: 10.1016/j.leukres.2017.07.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 06/24/2017] [Accepted: 07/23/2017] [Indexed: 10/19/2022]
Abstract
BACKGROUND Current results regarding treatment outcomes in acute myeloid leukemia (AML) point to significant differences between low- and middle-income countries (LMIC) and high-income countries (HIC). Excluding well-known socioeconomic issues, genetic markers important for prognosis have not been properly incorporated into the clinical practice so far and their usefulness outside of well-controlled clinical trials remain unknown. METHODS Here, we assessed the clinical significance of the European LeukemiaNet (ELN) recommendations in 196 consecutive patients with AML in a real-life setting. All patients were younger than 60 years of age (49% male) and treated with conventional chemotherapy for induction and consolidation in three Brazilian Institutions that well represent Brazilian geographic and socioeconomic diversity. FINDINGS Multivariable analysis showed that ELN recommendations had a slight association with complete remission achievement (odds ratio: 0.74, 95% confidence interval, CI: 0.53-1.01; P=0.06), but were independently associated with poor overall survival (OS) (hazard ratio, HR: 1.3, 95% CI: 1.1-1.54; P=0.002), disease-free survival (DFS) (HR: 1.42, 95% CI: 1.03-1.95; P=0.028) and event-free survival (EFS) (HR: 1.24, 95% CI: 1.06-1.47; P=0.007), considering initial leukocyte counts and age as confounders. ELN recommendations had no impact on cumulative incidence of relapse (P=0.09). INTERPRETATION Our results suggest that within the context of LMIC, the prognostic markers recommended by ELN may be useful to predict patient's clinical outcomes; however, the OS, DFS and EFS were shorter than the reported in Europe and US for the respective risk groups.
Collapse
Affiliation(s)
| | - Ana Flávia Tibúrcio Ribeiro
- Postgraduate Program in Pathology, Federal University of Minas Gerais, Belo Horizonte, Brazil; Hematology Unit, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Andre D Américo
- Department of Internal Medicine, Medical School of Ribeirao Preto, Ribeirao Preto, Brazil
| | - Felipe M Furtado
- Department of Internal Medicine, Medical School of Ribeirao Preto, Ribeirao Preto, Brazil
| | - Ana B Glória
- Hematology Unit, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Aleide S Lima
- Department of Genetics, Federal University of Pernambuco, Recife, Brazil
| | - Silvana M Santos
- Department of Clinical Pathology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Sandra G Xavier
- Department of Clinical Pathology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Evandro M Fagundes
- Hematology Unit, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Eduardo M Rego
- Department of Internal Medicine, Medical School of Ribeirao Preto, Ribeirao Preto, Brazil; Center for Cell Based Therapy, University of Sao Paulo, Ribeirao Preto, Brazil.
| |
Collapse
|
17
|
Huang JL, Liu W, Tian LH, Chai TT, Liu Y, Zhang F, Fu HY, Zhou HR, Shen JZ. Upregulation of long non-coding RNA MALAT-1 confers poor prognosis and influences cell proliferation and apoptosis in acute monocytic leukemia. Oncol Rep 2017; 38:1353-1362. [PMID: 28713913 PMCID: PMC5549035 DOI: 10.3892/or.2017.5802] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 06/09/2017] [Indexed: 12/30/2022] Open
Abstract
Metastasis-associated lung adenocarcinoma transcript 1 (MALAT-1), a long non-coding RNA, has been documented to be a new prognostic marker and gene regulator in several types of cancer, but its potential involvement in acute myeloid leukemia (AML) remains unclear. This study investigated the expression and functional role of MALAT-1 in AML. MALAT-1 expression was assessed by real-time quantitative PCR. After lentiviral-mediated MALAT-1 knockdown, the proliferation of AML cells was determined by CCK-8 and colony formation assays. Cell cycle progression and apoptosis were evaluated by flow cytometry and the expression of caspase-3, −8 and −9 was assessed by western blot analysis. We found that MALAT-1 expression in patients with acute monocytic leukemia (M5) was significantly increased when compared with that of healthy controls, and the overall survival of M5 patients with high MALAT-1 expression was markedly reduced when compared with the overall survival of patients with low MALAT-1 expression. The analysis of cellular experiments showed that MALAT-1 silencing decreased the proliferation of M5 cells (U-937 and THP-1), inhibited cell cycle progression and increased apoptosis. Taken together, these findings suggest that high MALAT-1 expression is closely associated with poor prognosis in M5 patients and may play a role in leukemia cell proliferation and apoptosis, and may serve as a promising theranostic marker.
Collapse
Affiliation(s)
- Jin-Long Huang
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Wei Liu
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Li-Hong Tian
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Ting-Ting Chai
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Yang Liu
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Feng Zhang
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Hai-Ying Fu
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Hua-Rong Zhou
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Jian-Zhen Shen
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
18
|
Krug U, Gale RP, Berdel WE, Müller-Tidow C, Stelljes M, Metzeler K, Sauerland MC, Hiddemann W, Büchner T. Therapy of older persons with acute myeloid leukaemia. Leuk Res 2017; 60:1-10. [PMID: 28618329 DOI: 10.1016/j.leukres.2017.05.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 05/24/2017] [Accepted: 05/27/2017] [Indexed: 12/20/2022]
Abstract
Most persons age≥60 y with acute myeloid leukaemia (AML) die from their disease. When interpreting clinical trials data from these persons one must be aware of substantial selection biases. Randomized trials of post-remission treatments can be performed upfront or after achieving defined landmarks. Both strategies have important limitations. Selection of the appropriate treatment is critical. Age, performance score, co-morbidities and frailty provide useful data to treatment selection. If an intensive remission induction therapy is appropriate, therapy with cytarabine and an anthracycline is the most common regimen. Non-intensive therapies consist of the hypo-methylating drugs azacitidine and decitabine, low-dose cytarabine and supportive care. Feasibility of doing an allotransplant in older persons with AML is increasing. However, only very few qualify. Results of cytogenetic testing are risk factor in young and old persons with AML. Adverse abnormalities are more frequent in older persons. Although data about the frequency of mutations in older persons with AML is increasing their prognostic impact is less clear than in younger subjects. Neither differences in the distribution of cytogenetic risk, mutations, nor differences in clinical risk factors between younger and older persons with AML completely explain the age-dependent outcome. Many drugs are in clinical development in older persons with AML. Their potential role in the treatment of older persons with AML remains to be defined.
Collapse
Affiliation(s)
- Utz Krug
- Klinikum Leverkusen, Department of Medicine 3, Am Gesundheitspark 11, 51375 Leverkusen, Germany.
| | - Robert Peter Gale
- Haematology Research Centre, Division of Experimental Medicine, Department of Medicine, Imperial College London, London, UK.
| | - Wolfgang E Berdel
- University Hospital Münster, Department of Medicine A, Albert-Schweitzer-Campus 1, Geb. A1, 48129 Münster, Germany.
| | - Carsten Müller-Tidow
- University Hospital Heidelberg, Department of Medicine V, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.
| | - Matthias Stelljes
- University Hospital Münster, Department of Medicine A, Albert-Schweitzer-Campus 1, Geb. A1, 48129 Münster, Germany.
| | - Klaus Metzeler
- University Hospital Großhadern, IIIrd Medical Department, Marchioninistraße 15, 81377 München, Germany.
| | - M Cristina Sauerland
- University of Münster, Institute of Biostatistics and Clinical Research, Schmeddingstr 56, 48149 Münster, Germany.
| | - Wolfgang Hiddemann
- University Hospital Großhadern, IIIrd Medical Department, Marchioninistraße 15, 81377 München, Germany.
| | - Thomas Büchner
- University Hospital Münster, Department of Medicine A, Albert-Schweitzer-Campus 1, Geb. A1, 48129 Münster, Germany
| |
Collapse
|
19
|
Thomas X, de Botton S, Chevret S, Caillot D, Raffoux E, Lemasle E, Marolleau JP, Berthon C, Pigneux A, Vey N, Reman O, Simon M, Recher C, Cahn JY, Hermine O, Castaigne S, Celli-Lebras K, Ifrah N, Preudhomme C, Terré C, Dombret H. Randomized Phase II Study of Clofarabine-Based Consolidation for Younger Adults With Acute Myeloid Leukemia in First Remission. J Clin Oncol 2017; 35:1223-1230. [PMID: 28221862 DOI: 10.1200/jco.2016.70.4551] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose To evaluate the efficacy and safety of a clofarabine-based combination (CLARA) versus conventional high-dose cytarabine (HDAC) as postremission chemotherapy in younger patients with acute myeloid leukemia (AML). Patients and Methods Patients age 18 to 59 years old with intermediate- or unfavorable-risk AML in first remission and no identified donor for allogeneic stem-cell transplantation (SCT) were eligible. Two hundred twenty-one patients were randomly assigned to receive three CLARA or three HDAC consolidation cycles. The primary end point was relapse-free survival (RFS). To handle the confounding effect of SCT that could occur in patients with late donor identification, hazard ratios (HRs) of events were adjusted on the time-dependent treatment × SCT interaction term. Results At 2 years, RFS was 58.5% (95% CI, 49% to 67%) in the CLARA arm and 46.5% (95% CI, 37% to 55%) in the HDAC arm. Overall, 110 patients (55 in each arm) received SCT in first remission. On the basis of a multivariable Cox-adjusted treatment × SCT interaction, the HR of CLARA over HDAC before or in absence of SCT was 0.65 (95% CI, 0.43 to 0.98; P = .041). In a sensitivity analysis, when patients who received SCT in first remission were censored at SCT time, 2-year RFS was 53.3% (95% CI, 39% to 66%) in the CLARA arm and 31.0% (95% CI, 19% to 43%) in the HDAC arm (HR, 0.63; 95% CI, 0.41 to 0.98; P = .043). Gain in RFS could be related to the lower cumulative incidence of relapse observed in the CLARA arm versus the HDAC arm (33.9% v 46.4% at 2 years, respectively; cause-specific HR, 0.61; 95% CI, 0.40 to 0.94; P = .025). CLARA cycles were associated with higher hematologic and nonhematologic toxicity than HDAC cycles. Conclusion These results suggest that CLARA might be considered as a new chemotherapy option in younger patients with AML in first remission.
Collapse
Affiliation(s)
- Xavier Thomas
- Xavier Thomas, Lyon-Sud University Hospital, Pierre Bénite; Stéphane de Botton, Gustave-Roussy Cancer Institute, Villejuif; Sylvie Chevret, Emmanuel Raffoux, and Hervé Dombret, Paris Saint-Louis University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), University Paris Diderot; Olivier Hermine, Paris Necker University Hospital, AP-HP, University Paris Descartes; Karine Celli-Lebras, Acute Leukemia French Association Coordination Office, Paris Saint-Louis University Hospital, Paris; Denis Caillot, Dijon University Hospital, Dijon; Emilie Lemasle, Henri-Becquerel Cancer Center, Rouen; Jean-Pierre Marolleau, Amiens University Hospital, Amiens; Céline Berthon, Claude Huriez University Hospital; Claude Preudhomme, University of Lille, Claude Huriez University Hospital, Institut National de la Santé et de la Recherche Médicale UMR-S 1172, Jean-Pierre Aubert Research Institute, Lille; Arnaud Pigneux, Bordeaux Haut-Leveque University Hospital, Pessac; Norbert Vey, Paoli-Calmette Cancer Institute, Marseille; Oumedaly Reman, George-Clémenceau University Hospital, Caen; Marc Simon, Valenciennes Hospital, Valenciennes; Christian Recher, Toulouse Cancer University Institute, Toulouse; Jean-Yves Cahn, Grenoble University Hospital, Grenoble; Sylvie Castaigne and Christine Terré, Versailles University Hospital, Versailles-Saint Quentin University, Le Chesnay; and Norbert Ifrah, Angers University Hospital, Angers, France
| | - Stéphane de Botton
- Xavier Thomas, Lyon-Sud University Hospital, Pierre Bénite; Stéphane de Botton, Gustave-Roussy Cancer Institute, Villejuif; Sylvie Chevret, Emmanuel Raffoux, and Hervé Dombret, Paris Saint-Louis University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), University Paris Diderot; Olivier Hermine, Paris Necker University Hospital, AP-HP, University Paris Descartes; Karine Celli-Lebras, Acute Leukemia French Association Coordination Office, Paris Saint-Louis University Hospital, Paris; Denis Caillot, Dijon University Hospital, Dijon; Emilie Lemasle, Henri-Becquerel Cancer Center, Rouen; Jean-Pierre Marolleau, Amiens University Hospital, Amiens; Céline Berthon, Claude Huriez University Hospital; Claude Preudhomme, University of Lille, Claude Huriez University Hospital, Institut National de la Santé et de la Recherche Médicale UMR-S 1172, Jean-Pierre Aubert Research Institute, Lille; Arnaud Pigneux, Bordeaux Haut-Leveque University Hospital, Pessac; Norbert Vey, Paoli-Calmette Cancer Institute, Marseille; Oumedaly Reman, George-Clémenceau University Hospital, Caen; Marc Simon, Valenciennes Hospital, Valenciennes; Christian Recher, Toulouse Cancer University Institute, Toulouse; Jean-Yves Cahn, Grenoble University Hospital, Grenoble; Sylvie Castaigne and Christine Terré, Versailles University Hospital, Versailles-Saint Quentin University, Le Chesnay; and Norbert Ifrah, Angers University Hospital, Angers, France
| | - Sylvie Chevret
- Xavier Thomas, Lyon-Sud University Hospital, Pierre Bénite; Stéphane de Botton, Gustave-Roussy Cancer Institute, Villejuif; Sylvie Chevret, Emmanuel Raffoux, and Hervé Dombret, Paris Saint-Louis University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), University Paris Diderot; Olivier Hermine, Paris Necker University Hospital, AP-HP, University Paris Descartes; Karine Celli-Lebras, Acute Leukemia French Association Coordination Office, Paris Saint-Louis University Hospital, Paris; Denis Caillot, Dijon University Hospital, Dijon; Emilie Lemasle, Henri-Becquerel Cancer Center, Rouen; Jean-Pierre Marolleau, Amiens University Hospital, Amiens; Céline Berthon, Claude Huriez University Hospital; Claude Preudhomme, University of Lille, Claude Huriez University Hospital, Institut National de la Santé et de la Recherche Médicale UMR-S 1172, Jean-Pierre Aubert Research Institute, Lille; Arnaud Pigneux, Bordeaux Haut-Leveque University Hospital, Pessac; Norbert Vey, Paoli-Calmette Cancer Institute, Marseille; Oumedaly Reman, George-Clémenceau University Hospital, Caen; Marc Simon, Valenciennes Hospital, Valenciennes; Christian Recher, Toulouse Cancer University Institute, Toulouse; Jean-Yves Cahn, Grenoble University Hospital, Grenoble; Sylvie Castaigne and Christine Terré, Versailles University Hospital, Versailles-Saint Quentin University, Le Chesnay; and Norbert Ifrah, Angers University Hospital, Angers, France
| | - Denis Caillot
- Xavier Thomas, Lyon-Sud University Hospital, Pierre Bénite; Stéphane de Botton, Gustave-Roussy Cancer Institute, Villejuif; Sylvie Chevret, Emmanuel Raffoux, and Hervé Dombret, Paris Saint-Louis University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), University Paris Diderot; Olivier Hermine, Paris Necker University Hospital, AP-HP, University Paris Descartes; Karine Celli-Lebras, Acute Leukemia French Association Coordination Office, Paris Saint-Louis University Hospital, Paris; Denis Caillot, Dijon University Hospital, Dijon; Emilie Lemasle, Henri-Becquerel Cancer Center, Rouen; Jean-Pierre Marolleau, Amiens University Hospital, Amiens; Céline Berthon, Claude Huriez University Hospital; Claude Preudhomme, University of Lille, Claude Huriez University Hospital, Institut National de la Santé et de la Recherche Médicale UMR-S 1172, Jean-Pierre Aubert Research Institute, Lille; Arnaud Pigneux, Bordeaux Haut-Leveque University Hospital, Pessac; Norbert Vey, Paoli-Calmette Cancer Institute, Marseille; Oumedaly Reman, George-Clémenceau University Hospital, Caen; Marc Simon, Valenciennes Hospital, Valenciennes; Christian Recher, Toulouse Cancer University Institute, Toulouse; Jean-Yves Cahn, Grenoble University Hospital, Grenoble; Sylvie Castaigne and Christine Terré, Versailles University Hospital, Versailles-Saint Quentin University, Le Chesnay; and Norbert Ifrah, Angers University Hospital, Angers, France
| | - Emmanuel Raffoux
- Xavier Thomas, Lyon-Sud University Hospital, Pierre Bénite; Stéphane de Botton, Gustave-Roussy Cancer Institute, Villejuif; Sylvie Chevret, Emmanuel Raffoux, and Hervé Dombret, Paris Saint-Louis University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), University Paris Diderot; Olivier Hermine, Paris Necker University Hospital, AP-HP, University Paris Descartes; Karine Celli-Lebras, Acute Leukemia French Association Coordination Office, Paris Saint-Louis University Hospital, Paris; Denis Caillot, Dijon University Hospital, Dijon; Emilie Lemasle, Henri-Becquerel Cancer Center, Rouen; Jean-Pierre Marolleau, Amiens University Hospital, Amiens; Céline Berthon, Claude Huriez University Hospital; Claude Preudhomme, University of Lille, Claude Huriez University Hospital, Institut National de la Santé et de la Recherche Médicale UMR-S 1172, Jean-Pierre Aubert Research Institute, Lille; Arnaud Pigneux, Bordeaux Haut-Leveque University Hospital, Pessac; Norbert Vey, Paoli-Calmette Cancer Institute, Marseille; Oumedaly Reman, George-Clémenceau University Hospital, Caen; Marc Simon, Valenciennes Hospital, Valenciennes; Christian Recher, Toulouse Cancer University Institute, Toulouse; Jean-Yves Cahn, Grenoble University Hospital, Grenoble; Sylvie Castaigne and Christine Terré, Versailles University Hospital, Versailles-Saint Quentin University, Le Chesnay; and Norbert Ifrah, Angers University Hospital, Angers, France
| | - Emilie Lemasle
- Xavier Thomas, Lyon-Sud University Hospital, Pierre Bénite; Stéphane de Botton, Gustave-Roussy Cancer Institute, Villejuif; Sylvie Chevret, Emmanuel Raffoux, and Hervé Dombret, Paris Saint-Louis University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), University Paris Diderot; Olivier Hermine, Paris Necker University Hospital, AP-HP, University Paris Descartes; Karine Celli-Lebras, Acute Leukemia French Association Coordination Office, Paris Saint-Louis University Hospital, Paris; Denis Caillot, Dijon University Hospital, Dijon; Emilie Lemasle, Henri-Becquerel Cancer Center, Rouen; Jean-Pierre Marolleau, Amiens University Hospital, Amiens; Céline Berthon, Claude Huriez University Hospital; Claude Preudhomme, University of Lille, Claude Huriez University Hospital, Institut National de la Santé et de la Recherche Médicale UMR-S 1172, Jean-Pierre Aubert Research Institute, Lille; Arnaud Pigneux, Bordeaux Haut-Leveque University Hospital, Pessac; Norbert Vey, Paoli-Calmette Cancer Institute, Marseille; Oumedaly Reman, George-Clémenceau University Hospital, Caen; Marc Simon, Valenciennes Hospital, Valenciennes; Christian Recher, Toulouse Cancer University Institute, Toulouse; Jean-Yves Cahn, Grenoble University Hospital, Grenoble; Sylvie Castaigne and Christine Terré, Versailles University Hospital, Versailles-Saint Quentin University, Le Chesnay; and Norbert Ifrah, Angers University Hospital, Angers, France
| | - Jean-Pierre Marolleau
- Xavier Thomas, Lyon-Sud University Hospital, Pierre Bénite; Stéphane de Botton, Gustave-Roussy Cancer Institute, Villejuif; Sylvie Chevret, Emmanuel Raffoux, and Hervé Dombret, Paris Saint-Louis University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), University Paris Diderot; Olivier Hermine, Paris Necker University Hospital, AP-HP, University Paris Descartes; Karine Celli-Lebras, Acute Leukemia French Association Coordination Office, Paris Saint-Louis University Hospital, Paris; Denis Caillot, Dijon University Hospital, Dijon; Emilie Lemasle, Henri-Becquerel Cancer Center, Rouen; Jean-Pierre Marolleau, Amiens University Hospital, Amiens; Céline Berthon, Claude Huriez University Hospital; Claude Preudhomme, University of Lille, Claude Huriez University Hospital, Institut National de la Santé et de la Recherche Médicale UMR-S 1172, Jean-Pierre Aubert Research Institute, Lille; Arnaud Pigneux, Bordeaux Haut-Leveque University Hospital, Pessac; Norbert Vey, Paoli-Calmette Cancer Institute, Marseille; Oumedaly Reman, George-Clémenceau University Hospital, Caen; Marc Simon, Valenciennes Hospital, Valenciennes; Christian Recher, Toulouse Cancer University Institute, Toulouse; Jean-Yves Cahn, Grenoble University Hospital, Grenoble; Sylvie Castaigne and Christine Terré, Versailles University Hospital, Versailles-Saint Quentin University, Le Chesnay; and Norbert Ifrah, Angers University Hospital, Angers, France
| | - Céline Berthon
- Xavier Thomas, Lyon-Sud University Hospital, Pierre Bénite; Stéphane de Botton, Gustave-Roussy Cancer Institute, Villejuif; Sylvie Chevret, Emmanuel Raffoux, and Hervé Dombret, Paris Saint-Louis University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), University Paris Diderot; Olivier Hermine, Paris Necker University Hospital, AP-HP, University Paris Descartes; Karine Celli-Lebras, Acute Leukemia French Association Coordination Office, Paris Saint-Louis University Hospital, Paris; Denis Caillot, Dijon University Hospital, Dijon; Emilie Lemasle, Henri-Becquerel Cancer Center, Rouen; Jean-Pierre Marolleau, Amiens University Hospital, Amiens; Céline Berthon, Claude Huriez University Hospital; Claude Preudhomme, University of Lille, Claude Huriez University Hospital, Institut National de la Santé et de la Recherche Médicale UMR-S 1172, Jean-Pierre Aubert Research Institute, Lille; Arnaud Pigneux, Bordeaux Haut-Leveque University Hospital, Pessac; Norbert Vey, Paoli-Calmette Cancer Institute, Marseille; Oumedaly Reman, George-Clémenceau University Hospital, Caen; Marc Simon, Valenciennes Hospital, Valenciennes; Christian Recher, Toulouse Cancer University Institute, Toulouse; Jean-Yves Cahn, Grenoble University Hospital, Grenoble; Sylvie Castaigne and Christine Terré, Versailles University Hospital, Versailles-Saint Quentin University, Le Chesnay; and Norbert Ifrah, Angers University Hospital, Angers, France
| | - Arnaud Pigneux
- Xavier Thomas, Lyon-Sud University Hospital, Pierre Bénite; Stéphane de Botton, Gustave-Roussy Cancer Institute, Villejuif; Sylvie Chevret, Emmanuel Raffoux, and Hervé Dombret, Paris Saint-Louis University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), University Paris Diderot; Olivier Hermine, Paris Necker University Hospital, AP-HP, University Paris Descartes; Karine Celli-Lebras, Acute Leukemia French Association Coordination Office, Paris Saint-Louis University Hospital, Paris; Denis Caillot, Dijon University Hospital, Dijon; Emilie Lemasle, Henri-Becquerel Cancer Center, Rouen; Jean-Pierre Marolleau, Amiens University Hospital, Amiens; Céline Berthon, Claude Huriez University Hospital; Claude Preudhomme, University of Lille, Claude Huriez University Hospital, Institut National de la Santé et de la Recherche Médicale UMR-S 1172, Jean-Pierre Aubert Research Institute, Lille; Arnaud Pigneux, Bordeaux Haut-Leveque University Hospital, Pessac; Norbert Vey, Paoli-Calmette Cancer Institute, Marseille; Oumedaly Reman, George-Clémenceau University Hospital, Caen; Marc Simon, Valenciennes Hospital, Valenciennes; Christian Recher, Toulouse Cancer University Institute, Toulouse; Jean-Yves Cahn, Grenoble University Hospital, Grenoble; Sylvie Castaigne and Christine Terré, Versailles University Hospital, Versailles-Saint Quentin University, Le Chesnay; and Norbert Ifrah, Angers University Hospital, Angers, France
| | - Norbert Vey
- Xavier Thomas, Lyon-Sud University Hospital, Pierre Bénite; Stéphane de Botton, Gustave-Roussy Cancer Institute, Villejuif; Sylvie Chevret, Emmanuel Raffoux, and Hervé Dombret, Paris Saint-Louis University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), University Paris Diderot; Olivier Hermine, Paris Necker University Hospital, AP-HP, University Paris Descartes; Karine Celli-Lebras, Acute Leukemia French Association Coordination Office, Paris Saint-Louis University Hospital, Paris; Denis Caillot, Dijon University Hospital, Dijon; Emilie Lemasle, Henri-Becquerel Cancer Center, Rouen; Jean-Pierre Marolleau, Amiens University Hospital, Amiens; Céline Berthon, Claude Huriez University Hospital; Claude Preudhomme, University of Lille, Claude Huriez University Hospital, Institut National de la Santé et de la Recherche Médicale UMR-S 1172, Jean-Pierre Aubert Research Institute, Lille; Arnaud Pigneux, Bordeaux Haut-Leveque University Hospital, Pessac; Norbert Vey, Paoli-Calmette Cancer Institute, Marseille; Oumedaly Reman, George-Clémenceau University Hospital, Caen; Marc Simon, Valenciennes Hospital, Valenciennes; Christian Recher, Toulouse Cancer University Institute, Toulouse; Jean-Yves Cahn, Grenoble University Hospital, Grenoble; Sylvie Castaigne and Christine Terré, Versailles University Hospital, Versailles-Saint Quentin University, Le Chesnay; and Norbert Ifrah, Angers University Hospital, Angers, France
| | - Oumedaly Reman
- Xavier Thomas, Lyon-Sud University Hospital, Pierre Bénite; Stéphane de Botton, Gustave-Roussy Cancer Institute, Villejuif; Sylvie Chevret, Emmanuel Raffoux, and Hervé Dombret, Paris Saint-Louis University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), University Paris Diderot; Olivier Hermine, Paris Necker University Hospital, AP-HP, University Paris Descartes; Karine Celli-Lebras, Acute Leukemia French Association Coordination Office, Paris Saint-Louis University Hospital, Paris; Denis Caillot, Dijon University Hospital, Dijon; Emilie Lemasle, Henri-Becquerel Cancer Center, Rouen; Jean-Pierre Marolleau, Amiens University Hospital, Amiens; Céline Berthon, Claude Huriez University Hospital; Claude Preudhomme, University of Lille, Claude Huriez University Hospital, Institut National de la Santé et de la Recherche Médicale UMR-S 1172, Jean-Pierre Aubert Research Institute, Lille; Arnaud Pigneux, Bordeaux Haut-Leveque University Hospital, Pessac; Norbert Vey, Paoli-Calmette Cancer Institute, Marseille; Oumedaly Reman, George-Clémenceau University Hospital, Caen; Marc Simon, Valenciennes Hospital, Valenciennes; Christian Recher, Toulouse Cancer University Institute, Toulouse; Jean-Yves Cahn, Grenoble University Hospital, Grenoble; Sylvie Castaigne and Christine Terré, Versailles University Hospital, Versailles-Saint Quentin University, Le Chesnay; and Norbert Ifrah, Angers University Hospital, Angers, France
| | - Marc Simon
- Xavier Thomas, Lyon-Sud University Hospital, Pierre Bénite; Stéphane de Botton, Gustave-Roussy Cancer Institute, Villejuif; Sylvie Chevret, Emmanuel Raffoux, and Hervé Dombret, Paris Saint-Louis University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), University Paris Diderot; Olivier Hermine, Paris Necker University Hospital, AP-HP, University Paris Descartes; Karine Celli-Lebras, Acute Leukemia French Association Coordination Office, Paris Saint-Louis University Hospital, Paris; Denis Caillot, Dijon University Hospital, Dijon; Emilie Lemasle, Henri-Becquerel Cancer Center, Rouen; Jean-Pierre Marolleau, Amiens University Hospital, Amiens; Céline Berthon, Claude Huriez University Hospital; Claude Preudhomme, University of Lille, Claude Huriez University Hospital, Institut National de la Santé et de la Recherche Médicale UMR-S 1172, Jean-Pierre Aubert Research Institute, Lille; Arnaud Pigneux, Bordeaux Haut-Leveque University Hospital, Pessac; Norbert Vey, Paoli-Calmette Cancer Institute, Marseille; Oumedaly Reman, George-Clémenceau University Hospital, Caen; Marc Simon, Valenciennes Hospital, Valenciennes; Christian Recher, Toulouse Cancer University Institute, Toulouse; Jean-Yves Cahn, Grenoble University Hospital, Grenoble; Sylvie Castaigne and Christine Terré, Versailles University Hospital, Versailles-Saint Quentin University, Le Chesnay; and Norbert Ifrah, Angers University Hospital, Angers, France
| | - Christian Recher
- Xavier Thomas, Lyon-Sud University Hospital, Pierre Bénite; Stéphane de Botton, Gustave-Roussy Cancer Institute, Villejuif; Sylvie Chevret, Emmanuel Raffoux, and Hervé Dombret, Paris Saint-Louis University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), University Paris Diderot; Olivier Hermine, Paris Necker University Hospital, AP-HP, University Paris Descartes; Karine Celli-Lebras, Acute Leukemia French Association Coordination Office, Paris Saint-Louis University Hospital, Paris; Denis Caillot, Dijon University Hospital, Dijon; Emilie Lemasle, Henri-Becquerel Cancer Center, Rouen; Jean-Pierre Marolleau, Amiens University Hospital, Amiens; Céline Berthon, Claude Huriez University Hospital; Claude Preudhomme, University of Lille, Claude Huriez University Hospital, Institut National de la Santé et de la Recherche Médicale UMR-S 1172, Jean-Pierre Aubert Research Institute, Lille; Arnaud Pigneux, Bordeaux Haut-Leveque University Hospital, Pessac; Norbert Vey, Paoli-Calmette Cancer Institute, Marseille; Oumedaly Reman, George-Clémenceau University Hospital, Caen; Marc Simon, Valenciennes Hospital, Valenciennes; Christian Recher, Toulouse Cancer University Institute, Toulouse; Jean-Yves Cahn, Grenoble University Hospital, Grenoble; Sylvie Castaigne and Christine Terré, Versailles University Hospital, Versailles-Saint Quentin University, Le Chesnay; and Norbert Ifrah, Angers University Hospital, Angers, France
| | - Jean-Yves Cahn
- Xavier Thomas, Lyon-Sud University Hospital, Pierre Bénite; Stéphane de Botton, Gustave-Roussy Cancer Institute, Villejuif; Sylvie Chevret, Emmanuel Raffoux, and Hervé Dombret, Paris Saint-Louis University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), University Paris Diderot; Olivier Hermine, Paris Necker University Hospital, AP-HP, University Paris Descartes; Karine Celli-Lebras, Acute Leukemia French Association Coordination Office, Paris Saint-Louis University Hospital, Paris; Denis Caillot, Dijon University Hospital, Dijon; Emilie Lemasle, Henri-Becquerel Cancer Center, Rouen; Jean-Pierre Marolleau, Amiens University Hospital, Amiens; Céline Berthon, Claude Huriez University Hospital; Claude Preudhomme, University of Lille, Claude Huriez University Hospital, Institut National de la Santé et de la Recherche Médicale UMR-S 1172, Jean-Pierre Aubert Research Institute, Lille; Arnaud Pigneux, Bordeaux Haut-Leveque University Hospital, Pessac; Norbert Vey, Paoli-Calmette Cancer Institute, Marseille; Oumedaly Reman, George-Clémenceau University Hospital, Caen; Marc Simon, Valenciennes Hospital, Valenciennes; Christian Recher, Toulouse Cancer University Institute, Toulouse; Jean-Yves Cahn, Grenoble University Hospital, Grenoble; Sylvie Castaigne and Christine Terré, Versailles University Hospital, Versailles-Saint Quentin University, Le Chesnay; and Norbert Ifrah, Angers University Hospital, Angers, France
| | - Olivier Hermine
- Xavier Thomas, Lyon-Sud University Hospital, Pierre Bénite; Stéphane de Botton, Gustave-Roussy Cancer Institute, Villejuif; Sylvie Chevret, Emmanuel Raffoux, and Hervé Dombret, Paris Saint-Louis University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), University Paris Diderot; Olivier Hermine, Paris Necker University Hospital, AP-HP, University Paris Descartes; Karine Celli-Lebras, Acute Leukemia French Association Coordination Office, Paris Saint-Louis University Hospital, Paris; Denis Caillot, Dijon University Hospital, Dijon; Emilie Lemasle, Henri-Becquerel Cancer Center, Rouen; Jean-Pierre Marolleau, Amiens University Hospital, Amiens; Céline Berthon, Claude Huriez University Hospital; Claude Preudhomme, University of Lille, Claude Huriez University Hospital, Institut National de la Santé et de la Recherche Médicale UMR-S 1172, Jean-Pierre Aubert Research Institute, Lille; Arnaud Pigneux, Bordeaux Haut-Leveque University Hospital, Pessac; Norbert Vey, Paoli-Calmette Cancer Institute, Marseille; Oumedaly Reman, George-Clémenceau University Hospital, Caen; Marc Simon, Valenciennes Hospital, Valenciennes; Christian Recher, Toulouse Cancer University Institute, Toulouse; Jean-Yves Cahn, Grenoble University Hospital, Grenoble; Sylvie Castaigne and Christine Terré, Versailles University Hospital, Versailles-Saint Quentin University, Le Chesnay; and Norbert Ifrah, Angers University Hospital, Angers, France
| | - Sylvie Castaigne
- Xavier Thomas, Lyon-Sud University Hospital, Pierre Bénite; Stéphane de Botton, Gustave-Roussy Cancer Institute, Villejuif; Sylvie Chevret, Emmanuel Raffoux, and Hervé Dombret, Paris Saint-Louis University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), University Paris Diderot; Olivier Hermine, Paris Necker University Hospital, AP-HP, University Paris Descartes; Karine Celli-Lebras, Acute Leukemia French Association Coordination Office, Paris Saint-Louis University Hospital, Paris; Denis Caillot, Dijon University Hospital, Dijon; Emilie Lemasle, Henri-Becquerel Cancer Center, Rouen; Jean-Pierre Marolleau, Amiens University Hospital, Amiens; Céline Berthon, Claude Huriez University Hospital; Claude Preudhomme, University of Lille, Claude Huriez University Hospital, Institut National de la Santé et de la Recherche Médicale UMR-S 1172, Jean-Pierre Aubert Research Institute, Lille; Arnaud Pigneux, Bordeaux Haut-Leveque University Hospital, Pessac; Norbert Vey, Paoli-Calmette Cancer Institute, Marseille; Oumedaly Reman, George-Clémenceau University Hospital, Caen; Marc Simon, Valenciennes Hospital, Valenciennes; Christian Recher, Toulouse Cancer University Institute, Toulouse; Jean-Yves Cahn, Grenoble University Hospital, Grenoble; Sylvie Castaigne and Christine Terré, Versailles University Hospital, Versailles-Saint Quentin University, Le Chesnay; and Norbert Ifrah, Angers University Hospital, Angers, France
| | - Karine Celli-Lebras
- Xavier Thomas, Lyon-Sud University Hospital, Pierre Bénite; Stéphane de Botton, Gustave-Roussy Cancer Institute, Villejuif; Sylvie Chevret, Emmanuel Raffoux, and Hervé Dombret, Paris Saint-Louis University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), University Paris Diderot; Olivier Hermine, Paris Necker University Hospital, AP-HP, University Paris Descartes; Karine Celli-Lebras, Acute Leukemia French Association Coordination Office, Paris Saint-Louis University Hospital, Paris; Denis Caillot, Dijon University Hospital, Dijon; Emilie Lemasle, Henri-Becquerel Cancer Center, Rouen; Jean-Pierre Marolleau, Amiens University Hospital, Amiens; Céline Berthon, Claude Huriez University Hospital; Claude Preudhomme, University of Lille, Claude Huriez University Hospital, Institut National de la Santé et de la Recherche Médicale UMR-S 1172, Jean-Pierre Aubert Research Institute, Lille; Arnaud Pigneux, Bordeaux Haut-Leveque University Hospital, Pessac; Norbert Vey, Paoli-Calmette Cancer Institute, Marseille; Oumedaly Reman, George-Clémenceau University Hospital, Caen; Marc Simon, Valenciennes Hospital, Valenciennes; Christian Recher, Toulouse Cancer University Institute, Toulouse; Jean-Yves Cahn, Grenoble University Hospital, Grenoble; Sylvie Castaigne and Christine Terré, Versailles University Hospital, Versailles-Saint Quentin University, Le Chesnay; and Norbert Ifrah, Angers University Hospital, Angers, France
| | - Norbert Ifrah
- Xavier Thomas, Lyon-Sud University Hospital, Pierre Bénite; Stéphane de Botton, Gustave-Roussy Cancer Institute, Villejuif; Sylvie Chevret, Emmanuel Raffoux, and Hervé Dombret, Paris Saint-Louis University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), University Paris Diderot; Olivier Hermine, Paris Necker University Hospital, AP-HP, University Paris Descartes; Karine Celli-Lebras, Acute Leukemia French Association Coordination Office, Paris Saint-Louis University Hospital, Paris; Denis Caillot, Dijon University Hospital, Dijon; Emilie Lemasle, Henri-Becquerel Cancer Center, Rouen; Jean-Pierre Marolleau, Amiens University Hospital, Amiens; Céline Berthon, Claude Huriez University Hospital; Claude Preudhomme, University of Lille, Claude Huriez University Hospital, Institut National de la Santé et de la Recherche Médicale UMR-S 1172, Jean-Pierre Aubert Research Institute, Lille; Arnaud Pigneux, Bordeaux Haut-Leveque University Hospital, Pessac; Norbert Vey, Paoli-Calmette Cancer Institute, Marseille; Oumedaly Reman, George-Clémenceau University Hospital, Caen; Marc Simon, Valenciennes Hospital, Valenciennes; Christian Recher, Toulouse Cancer University Institute, Toulouse; Jean-Yves Cahn, Grenoble University Hospital, Grenoble; Sylvie Castaigne and Christine Terré, Versailles University Hospital, Versailles-Saint Quentin University, Le Chesnay; and Norbert Ifrah, Angers University Hospital, Angers, France
| | - Claude Preudhomme
- Xavier Thomas, Lyon-Sud University Hospital, Pierre Bénite; Stéphane de Botton, Gustave-Roussy Cancer Institute, Villejuif; Sylvie Chevret, Emmanuel Raffoux, and Hervé Dombret, Paris Saint-Louis University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), University Paris Diderot; Olivier Hermine, Paris Necker University Hospital, AP-HP, University Paris Descartes; Karine Celli-Lebras, Acute Leukemia French Association Coordination Office, Paris Saint-Louis University Hospital, Paris; Denis Caillot, Dijon University Hospital, Dijon; Emilie Lemasle, Henri-Becquerel Cancer Center, Rouen; Jean-Pierre Marolleau, Amiens University Hospital, Amiens; Céline Berthon, Claude Huriez University Hospital; Claude Preudhomme, University of Lille, Claude Huriez University Hospital, Institut National de la Santé et de la Recherche Médicale UMR-S 1172, Jean-Pierre Aubert Research Institute, Lille; Arnaud Pigneux, Bordeaux Haut-Leveque University Hospital, Pessac; Norbert Vey, Paoli-Calmette Cancer Institute, Marseille; Oumedaly Reman, George-Clémenceau University Hospital, Caen; Marc Simon, Valenciennes Hospital, Valenciennes; Christian Recher, Toulouse Cancer University Institute, Toulouse; Jean-Yves Cahn, Grenoble University Hospital, Grenoble; Sylvie Castaigne and Christine Terré, Versailles University Hospital, Versailles-Saint Quentin University, Le Chesnay; and Norbert Ifrah, Angers University Hospital, Angers, France
| | - Christine Terré
- Xavier Thomas, Lyon-Sud University Hospital, Pierre Bénite; Stéphane de Botton, Gustave-Roussy Cancer Institute, Villejuif; Sylvie Chevret, Emmanuel Raffoux, and Hervé Dombret, Paris Saint-Louis University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), University Paris Diderot; Olivier Hermine, Paris Necker University Hospital, AP-HP, University Paris Descartes; Karine Celli-Lebras, Acute Leukemia French Association Coordination Office, Paris Saint-Louis University Hospital, Paris; Denis Caillot, Dijon University Hospital, Dijon; Emilie Lemasle, Henri-Becquerel Cancer Center, Rouen; Jean-Pierre Marolleau, Amiens University Hospital, Amiens; Céline Berthon, Claude Huriez University Hospital; Claude Preudhomme, University of Lille, Claude Huriez University Hospital, Institut National de la Santé et de la Recherche Médicale UMR-S 1172, Jean-Pierre Aubert Research Institute, Lille; Arnaud Pigneux, Bordeaux Haut-Leveque University Hospital, Pessac; Norbert Vey, Paoli-Calmette Cancer Institute, Marseille; Oumedaly Reman, George-Clémenceau University Hospital, Caen; Marc Simon, Valenciennes Hospital, Valenciennes; Christian Recher, Toulouse Cancer University Institute, Toulouse; Jean-Yves Cahn, Grenoble University Hospital, Grenoble; Sylvie Castaigne and Christine Terré, Versailles University Hospital, Versailles-Saint Quentin University, Le Chesnay; and Norbert Ifrah, Angers University Hospital, Angers, France
| | - Hervé Dombret
- Xavier Thomas, Lyon-Sud University Hospital, Pierre Bénite; Stéphane de Botton, Gustave-Roussy Cancer Institute, Villejuif; Sylvie Chevret, Emmanuel Raffoux, and Hervé Dombret, Paris Saint-Louis University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), University Paris Diderot; Olivier Hermine, Paris Necker University Hospital, AP-HP, University Paris Descartes; Karine Celli-Lebras, Acute Leukemia French Association Coordination Office, Paris Saint-Louis University Hospital, Paris; Denis Caillot, Dijon University Hospital, Dijon; Emilie Lemasle, Henri-Becquerel Cancer Center, Rouen; Jean-Pierre Marolleau, Amiens University Hospital, Amiens; Céline Berthon, Claude Huriez University Hospital; Claude Preudhomme, University of Lille, Claude Huriez University Hospital, Institut National de la Santé et de la Recherche Médicale UMR-S 1172, Jean-Pierre Aubert Research Institute, Lille; Arnaud Pigneux, Bordeaux Haut-Leveque University Hospital, Pessac; Norbert Vey, Paoli-Calmette Cancer Institute, Marseille; Oumedaly Reman, George-Clémenceau University Hospital, Caen; Marc Simon, Valenciennes Hospital, Valenciennes; Christian Recher, Toulouse Cancer University Institute, Toulouse; Jean-Yves Cahn, Grenoble University Hospital, Grenoble; Sylvie Castaigne and Christine Terré, Versailles University Hospital, Versailles-Saint Quentin University, Le Chesnay; and Norbert Ifrah, Angers University Hospital, Angers, France
| |
Collapse
|
20
|
Espirito Santo A, Chacim S, Ferreira I, Leite L, Moreira C, Pereira D, Dantas M, Nunes M, Viterbo L, Moreira I, Martins A, Oliveira I, Domingues N, Mariz J, Medeiros R. Southwestern Oncology Group pretreatment risk criteria as predictive or prognostic factors in acute myeloid leukemia. Mol Clin Oncol 2017; 6:384-388. [PMID: 28451418 DOI: 10.3892/mco.2017.1134] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 07/25/2017] [Indexed: 11/05/2022] Open
Abstract
Acute myeloid leukemia (AML) is a clonal hematological malignant condition and the implications of pretreatment risk criteria as predictive or prognostic factors are constantly under evaluation. With this study, the authors' intent was to characterize AML patients and to evaluate the clinical outcome associated with Southwestern Oncology Group (SWOG) coding pretreatment risk criteria/cytogenetic score. Between 2002 and 2010, 225 patients were diagnosed with AML at the Portuguese Institute of Oncology (Porto, Portugal). From this patient group, 128 patients aged <65 years were selected. The patients were treated using a combination of cytarabine and anthracycline, with the addition of cyclosporine when bone marrow dysplasia was observed. A median survival of 24 months was observed in this group. The patients were divided in subgroups according to the SWOG pretreatment risk criteria. We observed a statistically significant association of non-favorable SWOG coding with female gender [P=0.025; risk ratio (RR)=3.632, 95% confidence interval (CI): 1.113-11.852], indication for allogeneic bone marrow transplantation (P=0.023, RR=1.317, 95% CI: 1.184-1.465), complete response achievement (P=0.013, RR=1.385, 95% CI: 11.232-1.556) and relapse (P=0.048, RR=3.181, 95% CI: 10.966-10.478). Furthermore, SWOG pretreatment risk criteria also significantly affected global overall survival (OS; P=0.003) and OS at 5 years (P=0.001). A multivariate Cox regression analysis supported response to induction therapy (3-year OS: P=0.011, RR=0.385, 95% CI: 10.184-0.806; 5-year OS: P=0.012, RR=0.388, 95% CI: 10.597-1.994), consolidation (3-year OS: P=0.005, RR=0.328, 95% CI: 0.150-0.720; 5-year OS: P=0.002, RR=0.308, 95% CI: 0.144-0.657) and the diagnosis of therapy-related aml (3-year OS: P=0.016, RR=2.756, 95% CI: 0.486-1.281; 5-year OS: P=0.031, RR=2.369, 95% CI: 1.081-5.189) as prognostic factors, but this was not confirmed for SWOG pretreatment risk criteria. Therefore, we concluded that the reproducibility of the application of the SWOG pretreatment risk criteria may not be available as a prognostic factor in every acute leukemia population. However, its application as a predictive factor of response has been confirmed in our population.
Collapse
Affiliation(s)
- Ana Espirito Santo
- Department of Onco-Hematology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal.,Faculty of Medicine, University of Porto, 4000-286 Porto, Portugal
| | - Sergio Chacim
- Department of Onco-Hematology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal
| | - Isabel Ferreira
- Department of Onco-Hematology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal
| | - Luis Leite
- Department of Onco-Hematology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal
| | - Claudia Moreira
- Department of Onco-Hematology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal
| | - Dulcineia Pereira
- Department of Onco-Hematology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal
| | - Margarida Dantas
- Department of Onco-Hematology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal
| | - Marta Nunes
- Department of Onco-Hematology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal
| | - Luisa Viterbo
- Department of Onco-Hematology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal
| | - Ilidia Moreira
- Department of Onco-Hematology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal
| | - Angelo Martins
- Department of Onco-Hematology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal
| | - Isabel Oliveira
- Department of Onco-Hematology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal
| | - Nelson Domingues
- Department of Onco-Hematology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal
| | - Jose Mariz
- Department of Onco-Hematology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology Group-CI, Portuguese Institute of Oncology, 4200-072 Porto, Portugal.,Abel Salazar Institute of Biomedical Sciences, University of Porto, 4000-286 Porto, Portugal.,Oncology Department, Portuguese Institute of Oncology, 4200-072 Porto, Portugal.,Biomedical Research Center, Faculty of Health Sciences, Fernando Pessoa University, 4000-286 Porto, Portugal.,Research Department, Portuguese League Against Cancer, 4200-072 Porto, Portugal
| |
Collapse
|
21
|
Is there justification for 4 cycles of consolidation therapy in AML? Best Pract Res Clin Haematol 2016; 29:341-344. [DOI: 10.1016/j.beha.2016.10.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
22
|
Yang DQ, Zhou JD, Wang YX, Deng ZQ, Yang J, Yao DM, Qian Z, Yang L, Lin J, Qian J. LowmiR-34cexpression is associated with poor outcome inde novoacute myeloid leukemia. Int J Lab Hematol 2016; 39:42-50. [PMID: 27577964 DOI: 10.1111/ijlh.12566] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 07/12/2016] [Indexed: 12/30/2022]
Affiliation(s)
- D.-Q. Yang
- Department of Hematology; Affiliated People's Hospital of Jiangsu University; Zhenjiang Jiangsu China
| | - J.-D. Zhou
- Department of Hematology; Affiliated People's Hospital of Jiangsu University; Zhenjiang Jiangsu China
| | - Y.-X. Wang
- Department of Hematology; Affiliated People's Hospital of Jiangsu University; Zhenjiang Jiangsu China
| | - Z.-Q. Deng
- Laboratory Center; Affiliated People's Hospital of Jiangsu University; Zhenjiang Jiangsu China
| | - J. Yang
- Department of Hematology; Affiliated People's Hospital of Jiangsu University; Zhenjiang Jiangsu China
| | - D.-M. Yao
- Laboratory Center; Affiliated People's Hospital of Jiangsu University; Zhenjiang Jiangsu China
| | - Z. Qian
- Department of Hematology; Affiliated People's Hospital of Jiangsu University; Zhenjiang Jiangsu China
| | - L. Yang
- Department of Hematology; Affiliated People's Hospital of Jiangsu University; Zhenjiang Jiangsu China
| | - J. Lin
- Laboratory Center; Affiliated People's Hospital of Jiangsu University; Zhenjiang Jiangsu China
| | - J. Qian
- Department of Hematology; Affiliated People's Hospital of Jiangsu University; Zhenjiang Jiangsu China
| |
Collapse
|
23
|
Clinical outcomes of patients with acute myeloid leukemia: evaluation of genetic and molecular findings in a real-life setting. Blood 2015; 126:1863-5. [PMID: 26320101 DOI: 10.1182/blood-2015-07-657551] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
24
|
Chang TS, Tsai CK, Liang CS. Clozapine Use in Refractory Schizophrenia Comorbid With Acute Myeloid Leukemia. PSYCHOSOMATICS 2015; 56:302-5. [DOI: 10.1016/j.psym.2015.02.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 02/23/2015] [Accepted: 02/23/2015] [Indexed: 11/26/2022]
|
25
|
Bahl A, Sharma A, Raina V, Kumar L, Bakhshi S, Gupta R, Kumar R. Long-term outcomes for patients with acute myeloid leukemia: a single-center experience from AIIMS, India. Asia Pac J Clin Oncol 2015; 11:242-52. [PMID: 25639656 DOI: 10.1111/ajco.12333] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2014] [Indexed: 11/29/2022]
Abstract
AIM To analyze clinicopathological characteristics of acute myeloid leukemia (AML) patients and to evaluate long-term outcome of these patients presented to single tertiary care center in India. METHODS We evaluated outcomes of 480 patients (age 8-60 years), classified into good, intermediate and poor risk according to cytogenetic results. Standard "3 + 7" induction therapy with dose of daunorubicin ranging from 45 to 90 mg/m(2) followed by two to three courses of high-dose cytarabine (12-18 g/m(2) ) as consolidation therapy was given to majority. RESULTS The complete remission rate of the treated population (407 patients) was 70% with 84.8% in good risk, 67.9% in intermediate risk and 54.2% in poor risk (P = 0.0001). Induction mortality was 18.4%. One hundred twenty-nine patients relapsed with median treatment free interval of 10.4 months. At a median follow-up of 34.5 months, the median overall survival (OS) was 20.6 months with an estimated 5-year survival rate of 35.5%. No difference was found in OS between the three risk groups; however, patients with intermediate risk had a better leukemia-free survival (LFS) in comparison to good risk. Multivariate analysis showed age, performance status, treatment completion and hematopoietic stem cell transplant affecting OS, while only treatment completion affected LFS. CONCLUSION This is one of the largest single-center studies reflecting more accurately the outcome of AML in India. These results are likely due to uniform treatment protocols, intensification of induction and post-remission treatments with comprehensive supportive care.
Collapse
Affiliation(s)
- Ankur Bahl
- Department of Medical Oncology, Dr. B. R. A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Atul Sharma
- Department of Medical Oncology, Dr. B. R. A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Vinod Raina
- Department of Medical Oncology, Dr. B. R. A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Lalit Kumar
- Department of Medical Oncology, Dr. B. R. A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Sameer Bakhshi
- Department of Medical Oncology, Dr. B. R. A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Ritu Gupta
- Department of Laboratory Oncology, Dr. B. R. A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Rajeev Kumar
- Department of Laboratory Oncology, Dr. B. R. A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
26
|
Rohatiner AZS, Smith ML, Spinelli O, Rambaldi A, Bassan R, di Bona E, Rodeghiero F, Raimondi R, Björkholm M, Johnson S, Newland AC, Cavenagh JD, Macdougall F, Waters R, Fitzgibbon J, Barbui T, Lister A. Myeloblative therapy with autologous haematopoietic stem cell support as consolidation of first remission in acute myeloid leukaemia - very long follow-up. Br J Haematol 2014; 167:724-6. [PMID: 25074579 DOI: 10.1111/bjh.13055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
27
|
Overexpression of BAALC: clinical significance in Chinese de novo acute myeloid leukemia. Med Oncol 2014; 32:386. [PMID: 25428390 DOI: 10.1007/s12032-014-0386-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Accepted: 11/18/2014] [Indexed: 10/24/2022]
Abstract
To investigate the expression of brain and acute leukemia, cytoplasmic (BAALC) and analyze its clinical significance in Chinese de novo acute myeloid leukemia (AML). Real-time quantitative PCR (RQ-PCR) was carried out to detect BAALC transcript level in 121 de novo AML patients and 41 normal controls. BAALC transcript level in AML patients was significantly up-regulated compared with normal controls (P < 0.001). Patients with high BAALC expression had significantly older age than those with low BAALC expression (P = 0.021). The percentage of blasts in bone marrow of the BAALC high-expressed patients was significantly higher than that in the low-expressed patients (P < 0.001). The incidence of BAALC overexpression was significantly higher in M0/M1 (8/9, 89 %) and M2 subtypes (33/48, 68 %) than in M3 subtype (6/27, 22 %) (P < 0.001). The frequency of IDH1/2 wild type in CN-AML patients with high BAALC expression was significantly higher than those with low BAALC expression (P = 0.031). BAALC high-expressed patients had a significantly lower complete remission than low-expressed patients in both entire AML cohort and CN-AML (P = 0.013 and 0.029, respectively). Furthermore, both whole AML cohort and CN-AML patients with high BAALC expression showed a shorter overall survival than those with low BAALC expression (P = 0.002 and 0.008, respectively). Multivariate analysis confirmed high BAALC expression as an independent adverse prognostic factor in both AML and CN-AML patients. Our study indicates that overexpression of BAALC serves as an independent prognostic biomarker in both whole AML cohort and CN-AML patients.
Collapse
|
28
|
Li W, Gong X, Sun M, Zhao X, Gong B, Wei H, Mi Y, Wang J. High-dose cytarabine in acute myeloid leukemia treatment: a systematic review and meta-analysis. PLoS One 2014; 9:e110153. [PMID: 25299623 PMCID: PMC4192550 DOI: 10.1371/journal.pone.0110153] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 09/17/2014] [Indexed: 12/22/2022] Open
Abstract
The optimal dose, scheme, and clinical setting for Ara-C in acute myeloid leukemia (AML) treatment remain uncertain. In this study, we performed a meta-analysis to systematically assess the impact of high-dose cytarabine (HDAC) on AML therapy during the induction and consolidation stages. Twenty-two trials with a total of 5,945 de novo AML patients were included in the meta-analysis. Only patients less than 60 year-old were included in the study. Using HDAC in induction therapy was beneficial for RFS (HR = 0.57; 95% CI, 0.35-0.93; P = 0.02) but not so for CR rate (HR = 1.01; 95% CI, 0.93-1.09; P = 0.88) and OS (HR = 0.83; 95% CI, 0.66-1.03; P = 0.1). In consolidation therapy, HDAC showed significant RFS benefits (HR = 0.67; 95% CI, 0.49-0.9; P = 0.008) especially for the favorable-risk group (HR = 0.38; 95% CI, 0.21-0.69; P = 0.001) compared with SDAC (standard dose cytarabine), although no OS advantage was observed (HR = 0.84; 95% CI, 0.55-1.27; P = 0.41). HDAC treatment seemed less effective than auto-BMT/allo-BMT treatment (HR = 1.66, 95% CI, 1.3-2.14; P<0.0001) with similar OS. HDAC treatment led to lower relapse rate in induction and consolidation therapy than SDAC treatment, especially for the favorable-risk group. Auto-BMT/allo-BMT was more beneficial in prolonging RFS than HDAC.
Collapse
Affiliation(s)
- Wei Li
- Leukemia Diagnosis and Treatment Center, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union of Medical College, Tianjin, China
| | - Xiaoyuan Gong
- Leukemia Diagnosis and Treatment Center, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union of Medical College, Tianjin, China
| | - Mingyuan Sun
- Leukemia Diagnosis and Treatment Center, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union of Medical College, Tianjin, China
| | - Xingli Zhao
- Leukemia Diagnosis and Treatment Center, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union of Medical College, Tianjin, China
| | - Benfa Gong
- Leukemia Diagnosis and Treatment Center, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union of Medical College, Tianjin, China
| | - Hui Wei
- Leukemia Diagnosis and Treatment Center, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union of Medical College, Tianjin, China
| | - Yingchang Mi
- Leukemia Diagnosis and Treatment Center, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union of Medical College, Tianjin, China
| | - Jianxiang Wang
- Leukemia Diagnosis and Treatment Center, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union of Medical College, Tianjin, China
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union of Medical College, Tianjin, China
| |
Collapse
|
29
|
Chiaretti S, Gianfelici V, Ceglie G, Foà R. Genomic characterization of acute leukemias. Med Princ Pract 2014; 23:487-506. [PMID: 24968698 PMCID: PMC5586934 DOI: 10.1159/000362793] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Accepted: 04/10/2014] [Indexed: 01/09/2023] Open
Abstract
Over the past two decades, hematologic malignancies have been extensively evaluated due to the introduction of powerful technologies, such as conventional karyotyping, FISH analysis, gene and microRNA expression profiling, array comparative genomic hybridization and SNP arrays, and next-generation sequencing (including whole-exome sequencing and RNA-seq). These analyses have allowed for the refinement of the mechanisms underlying the leukemic transformation in several oncohematologic disorders and, more importantly, they have permitted the definition of novel prognostic algorithms aimed at stratifying patients at the onset of disease and, consequently, treating them in the most appropriate manner. Furthermore, the identification of specific molecular markers is opening the door to targeted and personalized medicine. The most important findings on novel acquisitions in the context of acute lymphoblastic leukemia of both B and T lineage and de novo acute myeloid leukemia are described in this review.
Collapse
Affiliation(s)
- Sabina Chiaretti
- Division of Hematology, Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy
| | | | | | | |
Collapse
|
30
|
Jabbour E, Ghanem H, Huang X, Ravandi F, Garcia-Manero G, O'Brien S, Faderl S, Pierce S, Choi S, Verstovsek S, Brandt M, Cortes J, Kantarjian H. Acute myeloid leukemia after myelodysplastic syndrome and failure of therapy with hypomethylating agents: an emerging entity with a poor prognosis. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2014; 14:93-97. [PMID: 24447728 PMCID: PMC4098769 DOI: 10.1016/j.clml.2013.10.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 10/21/2013] [Indexed: 01/10/2023]
Abstract
We assessed the outcomes of 63 patients with acute myeloid leukemia (AML) arising from myelodysplastic syndrome (MDS) after hypomethylating agent failure. Their median age was 63 years. All 63 patients had received ≥ 1 salvage regimens for AML, and 35 patients (55%) had received ≥ 2. Of the 31 patients (49%) who had received high-dose cytarabine (HDAC) at first relapse, 2 (6%) achieved complete remission (CR) and 4 (13%) CR with incomplete platelet recovery (overall response rate, 19%). Of the 32 patients (51%) who had received other treatments, including investigational agents, 4 (12%) achieved CR and 4 (12%) CR with incomplete platelet recovery (overall response rate, 24%). The median response duration was 20 weeks. With a median follow-up of 42 months from the AML diagnosis, the median survival (21 weeks) was similar between the 2 groups. The 1- and 2-year survival rate was 19% and 8%, respectively. Multivariate analysis identified low albumin, HDAC treatment, and platelet count < 50 × 10(9)/L as independent adverse factors for CR and a platelet count < 50 × 10(9)/L and age > 65 years as independent adverse factors for survival. Thus, the outcome of AML evolving from MDS after hypomethylating agent failure is poor and not improved with HDAC. Novel therapies directed toward this emerging entity are urgently needed.
Collapse
Affiliation(s)
- Elias Jabbour
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX.
| | - Hady Ghanem
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Xuelin Huang
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Farhad Ravandi
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Susan O'Brien
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Stephan Faderl
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sherry Pierce
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sangbum Choi
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Srdan Verstovsek
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Mark Brandt
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jorge Cortes
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Hagop Kantarjian
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
31
|
Warlick ED, Paulson K, Brazauskas R, Zhong X, Miller AM, Camitta BM, George B, Savani BN, Ustun C, Marks DI, Waller EK, Baron F, Freytes CO, Socie G, Akpek G, Schouten HC, Lazarus HM, Horwitz EM, Koreth J, Cahn JY, Bornhauser M, Seftel M, Cairo MS, Laughlin MJ, Sabloff M, Ringdén O, Gale RP, Kamble RT, Vij R, Gergis U, Mathews V, Saber W, Chen YB, Liesveld JL, Cutler CS, Ghobadi A, Uy GL, Eapen M, Weisdorf DJ, Litzow MR. Effect of postremission therapy before reduced-intensity conditioning allogeneic transplantation for acute myeloid leukemia in first complete remission. Biol Blood Marrow Transplant 2014; 20:202-8. [PMID: 24184335 PMCID: PMC3924751 DOI: 10.1016/j.bbmt.2013.10.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 10/28/2013] [Indexed: 11/23/2022]
Abstract
The impact of pretransplant (hematopoietic cell transplantation [HCT]) cytarabine consolidation therapy on post-HCT outcomes has yet to be evaluated after reduced-intensity or nonmyeloablative conditioning. We analyzed 604 adults with acute myeloid leukemia in first complete remission (CR1) reported to the Center for International Blood and Marrow Transplant Research who received a reduced-intensity or nonmyeloablative conditioning HCT from an HLA-identical sibling, HLA-matched unrelated donor, or umbilical cord blood donor from 2000 to 2010. We compared transplant outcomes based on exposure to cytarabine postremission consolidation. Three-year survival rates were 36% (95% confidence interval [CI], 29% to 43%) in the no consolidation arm and 42% (95% CI, 37% to 47%) in the cytarabine consolidation arm (P = .16). Disease-free survival was 34% (95% CI, 27% to 41%) and 41% (95% CI, 35% to 46%; P = .15), respectively. Three-year cumulative incidences of relapse were 37% (95% CI, 30% to 44%) and 38% (95% CI, 33% to 43%), respectively (P = .80). Multivariate regression confirmed no effect of consolidation on relapse, disease-free survival, and survival. Before reduced-intensity or nonmyeloablative conditioning HCT, these data suggest pre-HCT consolidation cytarabine does not significantly alter outcomes and support prompt transition to transplant as soon as morphologic CR1 is attained. If HCT is delayed while identifying a donor, our data suggest that consolidation does not increase transplant treatment-related mortality and is reasonable if required.
Collapse
Affiliation(s)
- Erica D Warlick
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota.
| | - Kristjan Paulson
- Department of Hematology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ruta Brazauskas
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Xiaobo Zhong
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Alan M Miller
- Department of Oncology, Baylor University Medical Center, Dallas, Texas
| | - Bruce M Camitta
- Department of Pediatrics, Midwest Center for Cancer and Blood Disorders, Medical College of Wisconsin and Children's Hospital of Wisconsin, Milwaukee, Wisconsin
| | - Biju George
- Department of Hematology, Christian Medical College Hospital, Vellore, India
| | - Bipin N Savani
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Celalettin Ustun
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - David I Marks
- Bristol Adult BMT Unit, Bristol Children's Hospital, Bristol, United Kingdom
| | - Edmund K Waller
- Bone Marrow and Stem Cell Transplant Center, Emory University Hospital, Atlanta, Georgia
| | - Frédéric Baron
- Universitaire de Liege, Centre Hospitalier Universitaire - Sart-Tilman, Liege, Belgium
| | - César O Freytes
- Department of Hematopoietic Stem Cell Transplant Program, South Texas Veterans Health Care System and University of Texas Health Science Center San Antonio, San Antonio, Texas
| | - Gérard Socie
- Service d'Hematologie, Hopital Saint Louis, Paris, France
| | - Gorgun Akpek
- SCTCT Program, Banner MD Anderson Cancer Center, Gilbert, Arizona
| | - Harry C Schouten
- Division of Hematology, Academische Ziekenhuis Maastricht, Maastricht, Netherlands
| | - Hillard M Lazarus
- Seidman Cancer Center, University Hospitals Case Medical Center, Cleveland, Ohio
| | - Edwin M Horwitz
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - John Koreth
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Jean-Yves Cahn
- Department of Hematology, University Hospital, Grenoble, France
| | - Martin Bornhauser
- Medizinische Klinik und Poliklinik I, Universitatsklinikum Carl Gustav Carus, Dresden, Germany
| | - Matthew Seftel
- Department of Hematology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Mitchell S Cairo
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, Maria Fareri Children's Hospital, New York Medical College, Valhalla, New York
| | - Mary J Laughlin
- Hematopoietic Cell Transplantation Program, University of Virginia, Charlottesville, Virginia
| | - Mitchell Sabloff
- Division of Hematology, Department of Medicine, University of Ottawa and The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Olle Ringdén
- Centre for Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Stockholm, Sweden
| | - Robert Peter Gale
- Section of Hematology, Division of Experimental Medicine, Department of Medicine, Imperial College, London, United Kingdom
| | - Rammurti T Kamble
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| | - Ravi Vij
- Washington University School of Medicine, St. Louis, Missouri
| | - Usama Gergis
- Weill Cornell Medical College, New York, New York
| | - Vikram Mathews
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Wael Saber
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Yi-Bin Chen
- Department of BMT, Massachusetts General Hospital, Boston, Massachusetts
| | - Jane L Liesveld
- Department of Hematology/Oncology, Strong Memorial Hospital, University of Rochester Medical Center, Rochester, New York
| | - Corey S Cutler
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Armin Ghobadi
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, Texas
| | - Geoffrey L Uy
- Section of Hematology, Division of Experimental Medicine, Department of Medicine, Imperial College, London, United Kingdom
| | - Mary Eapen
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Daniel J Weisdorf
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Mark R Litzow
- Department of Hematology and Internal Medicine, Mayo Clinic Rochester, Rochester, Minnesota
| |
Collapse
|
32
|
Willemze R, Suciu S, Meloni G, Labar B, Marie JP, Halkes CJ, Muus P, Mistrik M, Amadori S, Specchia G, Fabbiano F, Nobile F, Sborgia M, Camera A, Selleslag DL, Lefrère F, Magro D, Sica S, Cantore N, Beksac M, Berneman Z, Thomas X, Melillo L, Guimaraes JE, Leoni P, Luppi M, Mitra ME, Bron D, Fillet G, Marijt EW, Venditti A, Hagemeijer A, Mancini M, Jansen J, Cilloni D, Meert L, Fazi P, Vignetti M, Trisolini SM, Mandelli F, de Witte T. High-Dose Cytarabine in Induction Treatment Improves the Outcome of Adult Patients Younger Than Age 46 Years With Acute Myeloid Leukemia: Results of the EORTC-GIMEMA AML-12 Trial. J Clin Oncol 2014; 32:219-28. [DOI: 10.1200/jco.2013.51.8571] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Cytarabine plays a pivotal role in the treatment of patients with acute myeloid leukemia (AML). Most centers use 7 to 10 days of cytarabine at a daily dose of 100 to 200 mg/m2 for remission induction. Consensus has not been reached on the benefit of higher dosages of cytarabine. Patients and Methods The European Organisation for Research and Treatment of Cancer (EORTC) and Gruppo Italiano Malattie Ematologiche dell' Adulto (GIMEMA) Leukemia Groups conducted a randomized trial (AML-12; Combination Chemotherapy, Stem Cell Transplant and Interleukin-2 in Treating Patients With Acute Myeloid Leukemia) in 1,942 newly diagnosed patients with AML, age 15 to 60 years, comparing remission induction treatment containing daunorubicin, etoposide, and either standard-dose (SD) cytarabine (100 mg/m2 per day by continuous infusion for 10 days) or high-dose (HD) cytarabine (3,000 mg/m2 every 12 hours by 3-hour infusion on days 1, 3, 5, and 7). Patients in complete remission (CR) received a single consolidation cycle containing daunorubicin and intermediate-dose cytarabine (500 mg/m2 every 12 hours for 6 days). Subsequently, a stem-cell transplantation was planned. The primary end point was survival. Results At a median follow-up of 6 years, overall survival was 38.7% for patients randomly assigned to SD cytarabine and 42.5% for those randomly assigned to HD cytarabine (log-rank test P = .06; multivariable analysis P = .009). For patients younger than age 46 years, survival was 43.3% and 51.9%, respectively (P = .009; multivariable analysis P = .003), and for patients age 46 to 60 years, survival was 33.9% and 32.9%, respectively (P = .91). CR rates were 72.0% and 78.7%, respectively (P < .001) and were 75.6% and 82.4% for patients younger than age 46 years (P = .01) and 68.3% and 74.8% for patients age 46 years and older (P = .03). Patients of all ages with very-bad-risk cytogenetic abnormalities and/or FLT3-ITD (internal tandem duplication) mutation, or with secondary AML benefitted from HD cytarabine. Conclusion HD cytarabine produces higher remission and survival rates than SD cytarabine, especially in patients younger than age 46 years.
Collapse
Affiliation(s)
- Roelof Willemze
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Stefan Suciu
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Giovanna Meloni
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Boris Labar
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Jean-Pierre Marie
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Constantijn J.M. Halkes
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Petra Muus
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Martin Mistrik
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Sergio Amadori
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Giorgina Specchia
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Francesco Fabbiano
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Francesco Nobile
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Marco Sborgia
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Andrea Camera
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Dominik L.D. Selleslag
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Francois Lefrère
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Domenico Magro
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Simona Sica
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Nicola Cantore
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Meral Beksac
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Zwi Berneman
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Xavier Thomas
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Lorella Melillo
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Jose E. Guimaraes
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Pietro Leoni
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Mario Luppi
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Maria E. Mitra
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Dominique Bron
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Georges Fillet
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Erik W.A. Marijt
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Adriano Venditti
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Anne Hagemeijer
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Marco Mancini
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Joop Jansen
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Daniela Cilloni
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Liv Meert
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Paola Fazi
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Marco Vignetti
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Silvia M. Trisolini
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Franco Mandelli
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| | - Theo de Witte
- Roelof Willemze, Constantijn J.M. Halkes, and Erik W.A. Marijt, Leiden University Medical Center, Leiden; Petra Muus, Joop Jansen, and Theo de Witte, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; Stefan Suciu and Liv Meert, European Organisation for Research and Treatment of Cancer Headquarters; Dominique Bron, Hôpital Universitaire, Bordet-Erasme, Brussels; Dominik L.D. Selleslag, Algemeen Ziekenhuis Sint-Jan, Brugge; Zwi Berneman, Universitair Ziekenhuis, Antwerpen; Georges
| |
Collapse
|
33
|
Xu B, Chen G, Shi P, Guo X, Xiao P, Wang W, Zhou S. shRNA-Mediated BAALC knockdown affects proliferation and apoptosis in human acute myeloid leukemia cells. Hematology 2013; 17:35-40. [PMID: 22549446 DOI: 10.1179/102453312x13221316477499] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Affiliation(s)
- Bing Xu
- Department of HematologyNanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guoshu Chen
- Department of HematologyNanfang Hospital, Southern Medical University, Guangzhou, China
| | - Pengcheng Shi
- Department of HematologyNanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xutao Guo
- Department of HematologyNanfang Hospital, Southern Medical University, Guangzhou, China
| | - Pingnan Xiao
- Department of HematologyNanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weiguang Wang
- Research Institute in Healthcare Science, School of Applied Sciences, University of Wolverhampton, Wolverhampton, UK
| | - Shuyun Zhou
- Department of HematologyNanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
34
|
|
35
|
Lainey E, Sébert M, Thépot S, Scoazec M, Bouteloup C, Leroy C, De Botton S, Galluzzi L, Fenaux P, Kroemer G. Erlotinib antagonizes ABC transporters in acute myeloid leukemia. Cell Cycle 2012; 11:4079-92. [PMID: 23095522 DOI: 10.4161/cc.22382] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Erlotinib was originally developed as an epidermal growth factor receptor (EGFR)-specific inhibitor for the treatment of solid malignancies, yet also exerts significant EGFR-independent antileukemic effects in vitro and in vivo. The molecular mechanisms underlying the clinical antileukemic activity of erlotinib as a standalone agent have not yet been precisely elucidated. Conversely, in preclinical settings, erlotinib has been shown to inhibit the constitutive activation of SRC kinases and mTOR, as well as to synergize with the DNA methyltransferase inhibitor azacytidine (a reference therapeutic for a subset of leukemia patients) by promoting its intracellular accumulation. Here, we show that both erlotinib and gefitinib (another EGFR inhibitor) inhibit transmembrane transporters of the ATP-binding cassette (ABC) family, including P-glycoprotein (P-gp), multidrug resistance-associated proteins (MRPs) and breast cancer resistance protein (BCRP), also in acute myeloid leukemia (AML) cells that do not overexpress these pumps. Thus, inhibition of drug efflux by erlotinib and gefitinib selectively exacerbated (in a synergistic or additive fashion) the cytotoxic response of KG-1 cells to chemotherapeutic agents that are normally extruded by ABC transporters (e.g., doxorubicin and etoposide). Erlotinib limited drug export via ABC transporters by multiple mechanisms, including the downregulation of surface-exposed pumps and the modulation of their ATPase activity. The effects of erlotinib on drug efflux and its chemosensitization profile persisted in patient-derived CD34+ cells, suggesting that erlotinib might be particularly efficient in antagonizing leukemic (stem cell) subpopulations, irrespective of whether they exhibit or not increased drug efflux via ABC transporters.
Collapse
|
36
|
Detection of CD34, TdT, CD56, CD2, CD4, and CD14 by Flow Cytometry Is Associated With NPM1 and FLT3 Mutation Status in Cytogenetically Normal Acute Myeloid Leukemia. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2012; 12:274-9. [DOI: 10.1016/j.clml.2012.01.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Revised: 12/20/2011] [Accepted: 01/09/2012] [Indexed: 11/22/2022]
|
37
|
How J, Sykes J, Gupta V, Yee KWL, Schimmer AD, Schuh AC, Minden MD, Kamel-Reid S, Brandwein JM. Influence of FLT3-internal tandem duplication allele burden and white blood cell count on the outcome in patients with intermediate-risk karyotype acute myeloid leukemia. Cancer 2012; 118:6110-7. [PMID: 22736495 DOI: 10.1002/cncr.27683] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Revised: 03/06/2012] [Accepted: 03/27/2012] [Indexed: 11/07/2022]
Abstract
BACKGROUND In patients with acute myeloid leukemia (AML), testing for fms-like tyrosine kinase-3 (FLT3)-internal tandem duplication (FLT3-ITD) and nucleophosmin-1 (NPM1) mutations can allow for further prognostic subclassification, but less is known about the effects of FLT3-ITD allele burden and presenting white blood cell count (WBC) within molecular subgroups. METHODS The authors retrospectively assessed 206 adult patients who had AML with an intermediate-risk karyotype and who received treatment on a uniform induction and consolidation chemotherapy regimen. RESULTS The presenting WBC was a prognostic factor for survival only in patients who had an FLT3-ITD mutation. On multivariate analysis, after correcting for age, WBC, secondary AML, and blast percentage, nucleophosmin-1 (NPM1)-mutated/FLT3-ITD-negative patients had superior overall survival compared with patients in the other molecular subgroups. Patients who had FLT3-ITD mutations had an inferior overall survival compared with patients who had NPM1 wild-type/FLT3-negative disease, and patients who had low or intermediate levels of the FLT-ITD of mutant allele had overall and disease-free survival similar to those in patients who had high-level mutations. CONCLUSIONS NPM1 and FLT3-ITD status, age, WBC, and secondary AML were identified as important prognostic variables that can help to risk stratify patients with AML who have intermediate-risk cytogenetics. FLT3 allele burden had no significant influence on outcomes after correcting for other variables.
Collapse
Affiliation(s)
- Jonathan How
- Department of Medical Oncology and Hematology, Princess Margaret Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Heritable polymorphism predisposes to high BAALC expression in acute myeloid leukemia. Proc Natl Acad Sci U S A 2012; 109:6668-73. [PMID: 22493267 DOI: 10.1073/pnas.1203756109] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Overexpression of the brain and acute leukemia, cytoplasmic (BAALC) gene is implicated in myeloid leukemogenesis and associated with poor outcome in both acute myeloid leukemia (AML) and acute lymphoblastic leukemia patients. Additionally, high BAALC expression occurs in glioblastoma, melanoma, and childhood gastrointestinal stroma tumors, suggesting an oncogenic role for BAALC. However, the mechanisms underlying the deregulated expression are unknown. We hypothesized that a common heritable genetic feature located in cis might account for overexpression of BAALC in an allele-specific manner. By sequencing the genomic region of BAALC we identified nine informative single nucleotide polymorphisms (SNPs) and tested them for a possible association with BAALC expression levels. We show that BAALC overexpression occurs in the presence of the T allele of SNP rs62527607[GT], which creates a binding site for the activating RUNX1 transcription factor in the BAALC promoter region. The mechanism is demonstrated experimentally in vitro using luciferase reporter assays and electrophoretic mobility shift assay (EMSA) analysis. The association of high BAALC expression with the T allele and its correlations with RUNX1 expresser status are shown in vivo in a test set (n = 253) and validation set (n = 105) of samples from cytogenetically normal AML patients from different populations. Thus, we identify a heritable genomic feature predisposing to overexpression of an oncogene, thereby possibly leading to enhanced AML leukemogenesis. Our findings further suggest that genomic variants might become useful tools in the practice of personalized medicine.
Collapse
|
39
|
Cristóbal I, Garcia-Orti L, Cirauqui C, Cortes-Lavaud X, García-Sánchez MA, Calasanz MJ, Odero MD. Overexpression of SET is a recurrent event associated with poor outcome and contributes to protein phosphatase 2A inhibition in acute myeloid leukemia. Haematologica 2011; 97:543-50. [PMID: 22133779 DOI: 10.3324/haematol.2011.050542] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Protein phosphatase 2A is a novel potential therapeutic target in several types of chronic and acute leukemia, and its inhibition is a common event in acute myeloid leukemia. Upregulation of SET is essential to inhibit protein phosphatase 2A in chronic myeloid leukemia, but its importance in acute myeloid leukemia has not yet been explored. DESIGN AND METHODS We quantified SET expression by real time reverse transcriptase polymerase chain reaction in 214 acute myeloid leukemia patients at diagnosis. Western blot was performed in acute myeloid leukemia cell lines and in 16 patients' samples. We studied the effect of SET using cell viability assays. Bioinformatics analysis of the SET promoter, chromatin immunoprecipitation, and luciferase assays were performed to evaluate the transcriptional regulation of SET. RESULTS SET overexpression was found in 60/214 patients, for a prevalence of 28%. Patients with SET overexpression had worse overall survival (P<0.01) and event-free survival (P<0.01). Deregulation of SET was confirmed by western blot in both cell lines and patients' samples. Functional analysis showed that SET promotes proliferation, and restores cell viability after protein phosphatase 2A overexpression. We identified EVI1 overexpression as a mechanism involved in SET deregulation in acute myeloid leukemia cells. CONCLUSIONS These findings suggest that SET overexpression is a key mechanism in the inhibition of PP2A in acute myeloid leukemia, and that EVI1 overexpression contributes to the deregulation of SET. Furthermore, SET overexpression is associated with a poor outcome in acute myeloid leukemia, and it can be used to identify a subgroup of patients who could benefit from future treatments based on PP2A activators.
Collapse
Affiliation(s)
- Ion Cristóbal
- Division of Oncology, Center for Applied Medical Research (CIMA), University of Navarra, Pio XII-55 31008 Pamplona, Spain
| | | | | | | | | | | | | |
Collapse
|
40
|
Kumar CC. Genetic abnormalities and challenges in the treatment of acute myeloid leukemia. Genes Cancer 2011; 2:95-107. [PMID: 21779483 DOI: 10.1177/1947601911408076] [Citation(s) in RCA: 170] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Accepted: 03/17/2011] [Indexed: 01/31/2023] Open
Abstract
Acute myeloid leukemia (AML) is a hematopoietic disorder in which there are too many immature blood-forming cells accumulating in the bone marrow and interfering with the production of normal blood cells. It has long been recognized that AML is a clinically heterogeneous disease characterized by a multitude of chromosomal abnormalities and gene mutations, which translate to marked differences in responses and survival following chemotherapy. The cytogenetic and molecular genetic aberrations associated with AML are not mutually exclusive and often coexist in the leukemic cells. AML is a disease of the elderly, with a mean age of diagnosis of 70 years. Adverse cytogenetic abnormalities increase with age, and within each cytogenetic group, prognosis with standard treatment worsens with age. In the past 20 years, there has been little improvement in chemotherapeutic regimens and hence the overall survival for patients with AML. A huge unmet need exists for efficacious targeted therapies for elderly patients that are less toxic than available chemotherapy regimens. The multitude of chromosomal and genetic abnormalities makes the treatment of AML a challenging prospect. A detailed understanding of the molecular changes associated with the chromosomal and genetic abnormalities in AML is likely to provide a rationale for therapy design and biomarker development. This review summarizes the variety of cytogenetic and genetic changes observed in AML and gives an overview of the clinical status of new drugs in development.
Collapse
|
41
|
Novitzky N, Thomas V. In the absence of clinically significant graft vs. host disease, myeloablative conditioning may allow an effective graft vs. leukaemia effect. Leuk Res 2011; 36:104-9. [PMID: 21937110 DOI: 10.1016/j.leukres.2011.06.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2011] [Revised: 06/29/2011] [Accepted: 06/30/2011] [Indexed: 11/15/2022]
Abstract
In AML, prevention of GvHD leads to better tolerance of myeloablative therapy. 66 individuals with AML in CR underwent myeloablative conditioning and transplantation with allogeneic PBPC grafts. Median presentation age was 44.5 years. Karyotyping was intermediate in 48% and of unfavourable risk in 36%. For GvHD prophylaxis, PBPC harvests were incubated ex vivo with anti CD52 antibodies. TRM at day 100 and 1 year was 9% and 17%. At a median of 1018 days 65% are alive. Grade >1 GvHD was seen in 11%. GvHD and adverse karyotype were associated with treatment failure. In younger patients preservation of the dose intensity may improve cure rates.
Collapse
Affiliation(s)
- N Novitzky
- Leukaemia Unit and the Division of Haematology, Department Clinical Laboratory Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | | |
Collapse
|
42
|
Abstract
INTRODUCTION An increased understanding of cellular signaling pathways, like the JAK?STAT pathway, and the identification of the JAK2 V617F mutation in the classic Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs), has generated great interest in the development of targeted JAK2 inhibitors. In a recently completed Phase I?II study, ruxolitinib, a selective orally available JAK1 and JAK2 inhibitor, has shown efficacy in patients with advanced myelofibrosis. Constitutive activation of the JAK?STAT pathway has also been implicated in other hematological malignancies suggesting a potential role of JAK kinase inhibitors in these malignancies. AREAS COVERED This article reviews the chemistry, pharmacodynamics, pharmacokinetics, clinical efficacy, safety and tolerability of ruxolitinib. The literature for this article was retrieved from PubMed database searches using the keywords ?ruxolitinib?, ?INCB 018424?, ?JAK2 inhibitors? and ?leukemia?. EXPERT OPINION The JAK?STAT signaling pathway plays a vital role in leukemogenesis. Ruxolitinib, a potent JAK1 and JAK2 inhibitor, known to decrease spleen size and alleviate constitutional symptoms in myelofibrosis, represents a potentially promising agent for the treatment of leukemias by inhibiting the JAK?STAT signaling. Further studies of ruxolitinib, in patients with acute and chronic leukemias, are now needed to establish the clinical usefulness of this promising drug.
Collapse
Affiliation(s)
- Kiran Naqvi
- University of Texas M.D. Anderson Cancer Center, Department of Leukemia, Houston, TX, USA
| | - Srdan Verstovsek
- University of Texas M.D. Anderson Cancer Center, Department of Leukemia, Houston, TX, USA
| | - Hagop Kantarjian
- University of Texas M.D. Anderson Cancer Center, Department of Leukemia, Houston, TX, USA
| | - Farhad Ravandi
- University of Texas M.D. Anderson Cancer Center, Department of Leukemia, Houston, TX, USA
| |
Collapse
|
43
|
Canzoniero JV, Bhatnagar B, Baer MR, Gojo I. Upfront Therapy of Acute Myeloid Leukemia. Curr Oncol Rep 2011; 13:361-70. [DOI: 10.1007/s11912-011-0184-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
44
|
Comparison of high-dose cytarabine and timed-sequential chemotherapy as consolidation for younger adults with AML in first remission: the ALFA-9802 study. Blood 2011; 118:1754-62. [PMID: 21690555 DOI: 10.1182/blood-2011-04-349258] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
To assess the value of administering timed-sequential chemotherapy (TSC; 2 therapeutic sequences separated by a 4-day interval-free chemotherapy) or high-dose cytarabine (HDAraC) cycles in consolidation therapy for acute myeloid leukemia (AML), 459 patients 15 to 50 years of age were enrolled in the prospective randomized Acute Leukemia French Association-9802 trial. Complete remission was achieved in 89%. A total of 237 patients were then randomized to either TSC consolidation (120 patients) or HDAraC consolidation cycles (117 patients). Overall, there was no significant difference between the 2 consolidation arms (5-year event-free survival [EFS]: 41% for HDAraC vs 35% for TSC), or cumulative incidence of relapse, or treatment-related mortality. Cytogenetically normal AML NPM1(+) or CEBPA(+) and FLT3-ITD(-) had the same outcome as those with favorable cytogenetics. When considering favorable and unfavorable risk groups, the trend was in favor of HDAraC. However, the difference became significant when considering intermediate cytogenetics (5-year EFS: 49% vs 29%; P = .02), especially cytogenetically normal AML (5-year EFS: 48% vs 31%; P = .04), which was related to lower relapse rate and less toxicity. This study demonstrates that TSC did not produce any benefit when used as consolidation therapy in younger adults compared with HDAraC. This trial was registered at www.clinicaltrials.gov as #NCT00880243.
Collapse
|
45
|
MiR-100 regulates cell differentiation and survival by targeting RBSP3, a phosphatase-like tumor suppressor in acute myeloid leukemia. Oncogene 2011; 31:80-92. [PMID: 21643017 PMCID: PMC3253429 DOI: 10.1038/onc.2011.208] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Acute myeloblastic leukemia (AML) is characterized by the accumulation of abnormal myeloblasts (mainly granulocyte or monocyte precursors) in the bone marrow and blood. Though great progress has been made for improvement in clinical treatment during the past decades, only minority with AML achieve long-term survival. Therefore, further understanding mechanisms of leukemogenesis and exploring novel therapeutic strategies are still crucial for improving disease outcome. MicroRNA-100 (miR-100), a small non-coding RNA molecule, has been reported as a frequent event aberrantly expressed in patients with AML; however, the molecular basis for this phenotype and the statuses of its downstream targets have not yet been elucidated. In the present study, we found that the expression level of miR-100 in vivo was related to the stage of the maturation block underlying the subtypes of myeloid leukemia. In vitro experiments further demonstrated that miR-100 was required to promote the cell proliferation of promyelocytic blasts and arrest them differentiated to granulocyte/monocyte lineages. Significantly, we identified RBSP3, a phosphatase-like tumor suppressor, as a bona fide target of miR-100 and validated that RBSP3 was involved in cell differentiation and survival in AML. Moreover, we revealed a new pathway that miR-100 regulates G1/S transition and S-phase entry and blocks the terminal differentiation by targeting RBSP3, which partly in turn modulates the cell cycle effectors pRB/E2F1 in AML. These events promoted cell proliferation and blocked granulocyte/monocyte differentiation. Our data highlight an important role of miR-100 in the molecular etiology of AML, and implicate the potential application of miR-100 in cancer therapy.
Collapse
|
46
|
|
47
|
Individual patient data meta-analysis of randomized trials evaluating IL-2 monotherapy as remission maintenance therapy in acute myeloid leukemia. Blood 2011; 117:7007-13. [PMID: 21518931 DOI: 10.1182/blood-2011-02-337725] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
IL-2 is a natural, T cell-derived cytokine that stimulates the cytotoxic functions of T and natural killer cells. IL-2 monotherapy has been evaluated in several randomized clinical trials (RCTs) for remission maintenance in patients with acute myeloid leukemia (AML) in first complete remission (CR1), and none demonstrated a significant benefit of IL-2 monotherapy. The objective of this meta-analysis was to reliably determine IL-2 efficacy by combining all available individual patient data (IPD) from 5 RCTs (N = 905) and summary data from a sixth RCT (N = 550). Hazard ratios (HRs) were estimated using Cox regression models stratified by trial, with HR < 1 indicating treatment benefit. Combined IPD showed no benefit of IL-2 over no treatment in terms of leukemia-free survival (HR = 0.97; P = .74) or overall survival (HR = 1.08; P = .39). Analyses including the sixth RCT yielded qualitatively identical results (leukemia-free survival HR = 0.96, P = .52; overall survival HR = 1.06; P = .46). No significant heterogeneity was found between the trials. Prespecified subset analyses showed no interaction between the lack of IL-2 effect and any factor, including age, sex, baseline performance status, karyotype, AML subtype, and time from achievement of CR1 to initiation of maintenance therapy. We conclude that IL-2 alone is not an effective remission maintenance therapy for AML patients in CR1.
Collapse
|
48
|
Löwenberg B, Pabst T, Vellenga E, van Putten W, Schouten HC, Graux C, Ferrant A, Sonneveld P, Biemond BJ, Gratwohl A, de Greef GE, Verdonck LF, Schaafsma MR, Gregor M, Theobald M, Schanz U, Maertens J, Ossenkoppele GJ. Cytarabine dose for acute myeloid leukemia. N Engl J Med 2011; 364:1027-36. [PMID: 21410371 DOI: 10.1056/nejmoa1010222] [Citation(s) in RCA: 288] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Cytarabine (ara-C) is an important drug in the treatment of acute myeloid leukemia (AML). High-dose cytarabine (2000 to 3000 mg per square meter of body-surface area) is toxic but results in higher rates of relapse-free survival than does the conventional dose of 100 to 400 mg per square meter. Intermediate dose levels have not been thoroughly evaluated. METHODS We compared two induction regimens in patients 18 to 60 years of age (median, 49) who had newly diagnosed AML. The intermediate-dose group, totaling 431 patients, received cytarabine at a dose of 200 mg per square meter given by continuous intravenous infusion for 24 hours during cycle 1 of induction therapy and 1000 mg per square meter by infusion for 3 hours twice daily during cycle 2 of induction therapy. The high-dose group, totaling 429 patients, received a dose-escalated regimen of 1000 mg of cytarabine per square meter every 12 hours in cycle 1 and 2000 mg per square meter twice daily in cycle 2. Patients with a complete response did not receive additional cytarabine but received consolidation therapy in a third cycle of chemotherapy (mitoxantrone-etoposide) or underwent autologous or allogeneic stem-cell transplantation. Complete remission rates, survival rates, and toxic effects were assessed for each treatment group. RESULTS At a median follow-up of 5 years, no significant differences were noted between the intermediate-dose group and the high-dose group with respect to complete remission rates (80% and 82%, respectively), probability of relapse, event-free survival at 5 years (34% and 35%), or overall survival (40% and 42%). High-dose cytarabine provided no clear advantage in any prognostic subgroup. The high-dose treatment resulted in higher incidences of grade 3 and grade 4 toxic effects (in cycle 1), prolonged hospitalization, and delayed neutrophil recovery (in cycle 2) and platelet recovery (in cycles 2 and 3). CONCLUSIONS Induction therapy with cytarabine at the lower dose already produced maximal antileukemic effects for all response end points, suggesting a plateau in the dose-response relationship above this dose level. High-dose cytarabine results in excessive toxic effects without therapeutic benefit. (Netherlands Trial Register number, NTR230.).
Collapse
Affiliation(s)
- Bob Löwenberg
- Erasmus University Medical Center, Department of Hematology (L413), P.O. Box 2040, 3000 CA Rotterdam, the Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Autologous transplantation gives encouraging results for young adults with favorable-risk acute myeloid leukemia, but is not improved with gemtuzumab ozogamicin. Blood 2011; 117:5306-13. [PMID: 21415269 DOI: 10.1182/blood-2010-09-309229] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
We report the results of a prospective, randomized phase 3 trial evaluating the use of gemtuzumab ozogamicin (GO) in an intensive consolidation approach in 657 patients 17-60 years of age. Patients in first complete remission (CR1) after cytarabine and standard- or high-dose daunorubicin induction received 2 cycles of consolidation with high-dose cytarabine followed by peripheral blood progenitor cell collection. The 352 patients who entered consolidation were randomized to receive GO (n = 132) or not (n = 138) and then proceeded to autologous hematopoietic cell transplantation (HCT). GO was given to 67 patients. Median follow-up was 50.9 months. Results of the intention-to-treat analysis demonstrated a 4-year disease-free survival (DFS) of 33.6% versus 35.9% (P = .54) and an overall survival (OS) of 41.3% versus 41.9% (P = .52) for those randomized to receive GO versus no GO, respectively. Patients with favorable- and intermediate-risk acute myeloid leukemia (AML) treated with high-dose daunorubicin and autologous HCT had 4-year DFS rates of 60% and 40% and OS rates of 80% and 49.3%, respectively. For younger AML patients in CR1, autologous HCT should be considered in favorable- and intermediate-cytogenetic risk patients who do not have an allogeneic donor. The addition of a single dose of GO in this setting did not improve outcomes.
Collapse
|
50
|
|