51
|
Ilyas AM, Ahmad S, Faheem M, Naseer MI, Kumosani TA, Al-Qahtani MH, Gari M, Ahmed F. Next generation sequencing of acute myeloid leukemia: influencing prognosis. BMC Genomics 2015; 16 Suppl 1:S5. [PMID: 25924101 PMCID: PMC4315161 DOI: 10.1186/1471-2164-16-s1-s5] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Acute myeloid leukemia (AML) is a clonal disorder of the blood forming cells characterized by accumulation of immature blast cells in the bone marrow and peripheral blood. Being a heterogeneous disease, AML has been the subject of numerous studies that focus on unraveling the clinical, cellular and molecular variations with the aim to better understand and treat the disease. Cytogenetic-risk stratification of AML is well established and commonly used by clinicians in therapeutic management of cases with chromosomal abnormalities. Successive inclusion of novel molecular abnormalities has substantially modified the classification and understanding of AML in the past decade. With the advent of next generation sequencing (NGS) technologies the discovery of novel molecular abnormalities has accelerated. NGS has been successfully used in several studies and has provided an unprecedented overview of molecular aberrations as well as the underlying clonal evolution in AML. The extended spectrum of abnormalities discovered by NGS is currently under extensive validation for their prognostic and therapeutic values. In this review we highlight the recent advances in the understanding of AML in the NGS era.
Collapse
|
52
|
Choi Y, Lee JH, Seo EJ, Lee JH, Kim DY, Park CJ, Jang S, Cho YU, Seol M, Lee YS, Kang YA, Jeon M, Lee KH. Monosomal karyotype in acute myeloid leukemia and the role of allogeneic hematopoietic cell transplantation. Ann Hematol 2015; 94:795-801. [PMID: 25563594 DOI: 10.1007/s00277-014-2286-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 12/17/2014] [Indexed: 11/27/2022]
Abstract
Monosomal karyotype (MK) in acute myeloid leukemia (AML) is associated with an extremely poor outcome. The clinical significance of MK and the role of allogeneic hematopoietic cell transplantation (HCT) were evaluated in 749 Korean patients with newly diagnosed AML. MK was found in 9.3% of patients and was more frequent in patients with advanced age or secondary AML. Patients with MK had significantly lower blood leukocyte counts and bone marrow blast percentages, and they had lower complete remission (CR) rate (43%) and shorter median overall survival (OS) (6.5 months) and relapse-free survival (RFS) (10.0 months) than any other prognostic group. MK+ patients who received allogeneic HCT at the first CR had higher OS [hazard ratio (HR) 0.344, P = 0.018], RFS (HR 0.257, P = 0.006), and lower relapse probability (HR 0.264, P = 0.008) than those not receiving. This study's results confirmed poor outcomes for AML patients with MK and suggest that allogeneic HCT at the first CR may improve outcome.
Collapse
Affiliation(s)
- Yunsuk Choi
- Department of Hematology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro-43-gil, Songpa-gu, Seoul, 138-736, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
53
|
Sorror ML, Storb RF, Sandmaier BM, Maziarz RT, Pulsipher MA, Maris MB, Bhatia S, Ostronoff F, Deeg HJ, Syrjala KL, Estey E, Maloney DG, Appelbaum FR, Martin PJ, Storer BE. Comorbidity-age index: a clinical measure of biologic age before allogeneic hematopoietic cell transplantation. J Clin Oncol 2014; 32:3249-56. [PMID: 25154831 PMCID: PMC4178523 DOI: 10.1200/jco.2013.53.8157] [Citation(s) in RCA: 295] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
PURPOSE Age has long been used as a major factor for assessing suitability for allogeneic hematopoietic cell transplantation (HCT). The HCT-comorbidity index (HCT-CI) was developed as a measure of health status to predict mortality risk after HCT. Whether age, comorbidities, or both should guide decision making for HCT is unknown. PATIENTS AND METHODS Data from 3,033 consecutive recipients of HLA-matched grafts from five institutions contributed to this analysis. Patients were randomly divided into a training set to develop weights for age intervals and a validation set to assess the performance of prognostic models. RESULTS In the training set, patients age 20 to 39 years, 40 to 49 years, 50 to 59 years, and ≥ 60 years had hazard ratios for nonrelapse mortality (NRM) of 1.21 (P = .29), 1.48 (P = .04), 1.75 (P = .004), and 1.84 (P = .005), respectively, compared with those age younger than 20 years. Consequently, age ≥ 40 years was assigned a weight of 1 to be added to the HCT-CI to constitute a composite comorbidity/age index. In the validation set, the composite comorbidity/age score had statistically significantly higher c-statistic estimates for prediction of NRM (0.664 v 0.556; P < .001) and survival (0.682 v 0.560; P < .001) compared with age, respectively. Patients with comorbidity/age scores of 0 to 2 had comparable mortality risks regardless of conditioning regimens. Patients with scores of 3 to 4 and ≥ 5 had statistically significant higher mortality risks after high-dose versus nonmyeloablative regimens. CONCLUSION Age is a poor prognostic factor. The proposed composite measure allows integration of both comorbidities and age into clinical decision making and comparative-effectiveness research of HCT.
Collapse
Affiliation(s)
- Mohamed L Sorror
- Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, Paul J. Martin, and Barry E. Storer, Fred Hutchinson Cancer Research Center; Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, and Paul J. Martin, University of Washington School of Medicine; Barry E. Storer, University of Washington School of Public Health, Seattle, WA; Smita Bhatia, City of Hope School of Medicine, Duarte, CA; Richard T. Maziarz, Center for Hematologic Malignancies, Oregon Health & Science University Knight Cancer Institute and School of Medicine, Portland, OR; Michael A. Pulsipher, University of Utah School of Medicine and Primary Children's Medical Center, Huntsman Cancer Institute, Salt Lake City, UT; and Michael B. Maris, Colorado Blood Cancer Institute School of Medicine, Denver, CO.
| | - Rainer F Storb
- Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, Paul J. Martin, and Barry E. Storer, Fred Hutchinson Cancer Research Center; Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, and Paul J. Martin, University of Washington School of Medicine; Barry E. Storer, University of Washington School of Public Health, Seattle, WA; Smita Bhatia, City of Hope School of Medicine, Duarte, CA; Richard T. Maziarz, Center for Hematologic Malignancies, Oregon Health & Science University Knight Cancer Institute and School of Medicine, Portland, OR; Michael A. Pulsipher, University of Utah School of Medicine and Primary Children's Medical Center, Huntsman Cancer Institute, Salt Lake City, UT; and Michael B. Maris, Colorado Blood Cancer Institute School of Medicine, Denver, CO
| | - Brenda M Sandmaier
- Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, Paul J. Martin, and Barry E. Storer, Fred Hutchinson Cancer Research Center; Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, and Paul J. Martin, University of Washington School of Medicine; Barry E. Storer, University of Washington School of Public Health, Seattle, WA; Smita Bhatia, City of Hope School of Medicine, Duarte, CA; Richard T. Maziarz, Center for Hematologic Malignancies, Oregon Health & Science University Knight Cancer Institute and School of Medicine, Portland, OR; Michael A. Pulsipher, University of Utah School of Medicine and Primary Children's Medical Center, Huntsman Cancer Institute, Salt Lake City, UT; and Michael B. Maris, Colorado Blood Cancer Institute School of Medicine, Denver, CO
| | - Richard T Maziarz
- Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, Paul J. Martin, and Barry E. Storer, Fred Hutchinson Cancer Research Center; Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, and Paul J. Martin, University of Washington School of Medicine; Barry E. Storer, University of Washington School of Public Health, Seattle, WA; Smita Bhatia, City of Hope School of Medicine, Duarte, CA; Richard T. Maziarz, Center for Hematologic Malignancies, Oregon Health & Science University Knight Cancer Institute and School of Medicine, Portland, OR; Michael A. Pulsipher, University of Utah School of Medicine and Primary Children's Medical Center, Huntsman Cancer Institute, Salt Lake City, UT; and Michael B. Maris, Colorado Blood Cancer Institute School of Medicine, Denver, CO
| | - Michael A Pulsipher
- Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, Paul J. Martin, and Barry E. Storer, Fred Hutchinson Cancer Research Center; Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, and Paul J. Martin, University of Washington School of Medicine; Barry E. Storer, University of Washington School of Public Health, Seattle, WA; Smita Bhatia, City of Hope School of Medicine, Duarte, CA; Richard T. Maziarz, Center for Hematologic Malignancies, Oregon Health & Science University Knight Cancer Institute and School of Medicine, Portland, OR; Michael A. Pulsipher, University of Utah School of Medicine and Primary Children's Medical Center, Huntsman Cancer Institute, Salt Lake City, UT; and Michael B. Maris, Colorado Blood Cancer Institute School of Medicine, Denver, CO
| | - Michael B Maris
- Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, Paul J. Martin, and Barry E. Storer, Fred Hutchinson Cancer Research Center; Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, and Paul J. Martin, University of Washington School of Medicine; Barry E. Storer, University of Washington School of Public Health, Seattle, WA; Smita Bhatia, City of Hope School of Medicine, Duarte, CA; Richard T. Maziarz, Center for Hematologic Malignancies, Oregon Health & Science University Knight Cancer Institute and School of Medicine, Portland, OR; Michael A. Pulsipher, University of Utah School of Medicine and Primary Children's Medical Center, Huntsman Cancer Institute, Salt Lake City, UT; and Michael B. Maris, Colorado Blood Cancer Institute School of Medicine, Denver, CO
| | - Smita Bhatia
- Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, Paul J. Martin, and Barry E. Storer, Fred Hutchinson Cancer Research Center; Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, and Paul J. Martin, University of Washington School of Medicine; Barry E. Storer, University of Washington School of Public Health, Seattle, WA; Smita Bhatia, City of Hope School of Medicine, Duarte, CA; Richard T. Maziarz, Center for Hematologic Malignancies, Oregon Health & Science University Knight Cancer Institute and School of Medicine, Portland, OR; Michael A. Pulsipher, University of Utah School of Medicine and Primary Children's Medical Center, Huntsman Cancer Institute, Salt Lake City, UT; and Michael B. Maris, Colorado Blood Cancer Institute School of Medicine, Denver, CO
| | - Fabiana Ostronoff
- Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, Paul J. Martin, and Barry E. Storer, Fred Hutchinson Cancer Research Center; Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, and Paul J. Martin, University of Washington School of Medicine; Barry E. Storer, University of Washington School of Public Health, Seattle, WA; Smita Bhatia, City of Hope School of Medicine, Duarte, CA; Richard T. Maziarz, Center for Hematologic Malignancies, Oregon Health & Science University Knight Cancer Institute and School of Medicine, Portland, OR; Michael A. Pulsipher, University of Utah School of Medicine and Primary Children's Medical Center, Huntsman Cancer Institute, Salt Lake City, UT; and Michael B. Maris, Colorado Blood Cancer Institute School of Medicine, Denver, CO
| | - H Joachim Deeg
- Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, Paul J. Martin, and Barry E. Storer, Fred Hutchinson Cancer Research Center; Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, and Paul J. Martin, University of Washington School of Medicine; Barry E. Storer, University of Washington School of Public Health, Seattle, WA; Smita Bhatia, City of Hope School of Medicine, Duarte, CA; Richard T. Maziarz, Center for Hematologic Malignancies, Oregon Health & Science University Knight Cancer Institute and School of Medicine, Portland, OR; Michael A. Pulsipher, University of Utah School of Medicine and Primary Children's Medical Center, Huntsman Cancer Institute, Salt Lake City, UT; and Michael B. Maris, Colorado Blood Cancer Institute School of Medicine, Denver, CO
| | - Karen L Syrjala
- Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, Paul J. Martin, and Barry E. Storer, Fred Hutchinson Cancer Research Center; Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, and Paul J. Martin, University of Washington School of Medicine; Barry E. Storer, University of Washington School of Public Health, Seattle, WA; Smita Bhatia, City of Hope School of Medicine, Duarte, CA; Richard T. Maziarz, Center for Hematologic Malignancies, Oregon Health & Science University Knight Cancer Institute and School of Medicine, Portland, OR; Michael A. Pulsipher, University of Utah School of Medicine and Primary Children's Medical Center, Huntsman Cancer Institute, Salt Lake City, UT; and Michael B. Maris, Colorado Blood Cancer Institute School of Medicine, Denver, CO
| | - Elihu Estey
- Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, Paul J. Martin, and Barry E. Storer, Fred Hutchinson Cancer Research Center; Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, and Paul J. Martin, University of Washington School of Medicine; Barry E. Storer, University of Washington School of Public Health, Seattle, WA; Smita Bhatia, City of Hope School of Medicine, Duarte, CA; Richard T. Maziarz, Center for Hematologic Malignancies, Oregon Health & Science University Knight Cancer Institute and School of Medicine, Portland, OR; Michael A. Pulsipher, University of Utah School of Medicine and Primary Children's Medical Center, Huntsman Cancer Institute, Salt Lake City, UT; and Michael B. Maris, Colorado Blood Cancer Institute School of Medicine, Denver, CO
| | - David G Maloney
- Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, Paul J. Martin, and Barry E. Storer, Fred Hutchinson Cancer Research Center; Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, and Paul J. Martin, University of Washington School of Medicine; Barry E. Storer, University of Washington School of Public Health, Seattle, WA; Smita Bhatia, City of Hope School of Medicine, Duarte, CA; Richard T. Maziarz, Center for Hematologic Malignancies, Oregon Health & Science University Knight Cancer Institute and School of Medicine, Portland, OR; Michael A. Pulsipher, University of Utah School of Medicine and Primary Children's Medical Center, Huntsman Cancer Institute, Salt Lake City, UT; and Michael B. Maris, Colorado Blood Cancer Institute School of Medicine, Denver, CO
| | - Frederick R Appelbaum
- Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, Paul J. Martin, and Barry E. Storer, Fred Hutchinson Cancer Research Center; Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, and Paul J. Martin, University of Washington School of Medicine; Barry E. Storer, University of Washington School of Public Health, Seattle, WA; Smita Bhatia, City of Hope School of Medicine, Duarte, CA; Richard T. Maziarz, Center for Hematologic Malignancies, Oregon Health & Science University Knight Cancer Institute and School of Medicine, Portland, OR; Michael A. Pulsipher, University of Utah School of Medicine and Primary Children's Medical Center, Huntsman Cancer Institute, Salt Lake City, UT; and Michael B. Maris, Colorado Blood Cancer Institute School of Medicine, Denver, CO
| | - Paul J Martin
- Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, Paul J. Martin, and Barry E. Storer, Fred Hutchinson Cancer Research Center; Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, and Paul J. Martin, University of Washington School of Medicine; Barry E. Storer, University of Washington School of Public Health, Seattle, WA; Smita Bhatia, City of Hope School of Medicine, Duarte, CA; Richard T. Maziarz, Center for Hematologic Malignancies, Oregon Health & Science University Knight Cancer Institute and School of Medicine, Portland, OR; Michael A. Pulsipher, University of Utah School of Medicine and Primary Children's Medical Center, Huntsman Cancer Institute, Salt Lake City, UT; and Michael B. Maris, Colorado Blood Cancer Institute School of Medicine, Denver, CO
| | - Barry E Storer
- Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, Paul J. Martin, and Barry E. Storer, Fred Hutchinson Cancer Research Center; Mohamed L. Sorror, Rainer F. Storb, Brenda M. Sandmaier, Fabiana Ostronoff, H. Joachim Deeg, Karen L. Syrjala, Elihu Estey, David G. Maloney, Frederick R. Appelbaum, and Paul J. Martin, University of Washington School of Medicine; Barry E. Storer, University of Washington School of Public Health, Seattle, WA; Smita Bhatia, City of Hope School of Medicine, Duarte, CA; Richard T. Maziarz, Center for Hematologic Malignancies, Oregon Health & Science University Knight Cancer Institute and School of Medicine, Portland, OR; Michael A. Pulsipher, University of Utah School of Medicine and Primary Children's Medical Center, Huntsman Cancer Institute, Salt Lake City, UT; and Michael B. Maris, Colorado Blood Cancer Institute School of Medicine, Denver, CO
| |
Collapse
|
54
|
Abdel Hamid TM, El Gammal MM, Eibead GT, Saber MM, Abol Elazm OM. Clinical impact of SNP of P53 genes pathway on the adult AML patients. ACTA ACUST UNITED AC 2014; 20:328-35. [PMID: 25232917 DOI: 10.1179/1607845414y.0000000200] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
INTRODUCTION Acute myeloid leukemia (AML) is a highly heterogeneous disease, with biologically and prognostically different subtypes. AIM To study the impact of p53, p21, and mdm2 gene polymorphisms on the clinical outcome in adult AML patients treated at the National Cancer Institute (NCI) - Cairo University. METHODS Forty-eight adult AML patients presented to the Medical Oncology Department, NCI, from April 2010 till November 2011. Clinical data and bone marrow samples were obtained. Molecular genetic analysis involving P53, MDM2, and P21 single-nucleotide gene polymorphisms was done using polymerase chain reaction-restriction fragment length polymorphism coupled analysis. RESULTS The mean age was 35.7 years. After a median follow-up period of 12 months, 28 patients (58.4%) achieved complete remission (CR) and the overall survival (OS) was 8.7 months. Patients with homozygous Arg/arg at codon 72 of P53 had a better median OS months than Arg/Pro and Pro/Pro (13.4 vs. 8.4 vs. 1.5 months, respectively; P = 0.045). P53/p21 combination had a better median OS and disease-free survival (DFS) of 12.1 and 13.7 months for wild type cases (GG + Ser/ser) and 20.3 and 20.7 months for patients with either variant genes (GC + Ser/arg) compared with 1.1 and 1.9 months for patients with both variant genes (CC + arg/arg), (P = 0.037 and 0.004). The presence of wild genotype of either P21 or MDM2 may abolish the effect of P53 homozygous variant genotype on the OS. Neither p21nor mdm2 polymorphism alone showed an impact on OS or DFS. CR was not affected by any of the three gene polymorphisms. CONCLUSION The p53 pathway gene polymorphisms may affect the OS of adult AML patients.
Collapse
|
55
|
Wetzel D, Mueller BU, Mansouri Taleghani B, Baerlocher GM, Seipel K, Leibundgut K, Pabst T. Delayed Haematological recovery after autologous stem cell transplantation is associated with favourable outcome in acute myeloid leukaemia. Br J Haematol 2014; 168:268-73. [DOI: 10.1111/bjh.13118] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 07/29/2014] [Indexed: 11/29/2022]
Affiliation(s)
- Dana Wetzel
- Department of Medical Oncology; University Hospital and University of Berne; Berne Switzerland
| | - Beatrice U. Mueller
- Department of Clinical Research; University Hospital and University of Berne; Berne Switzerland
| | | | - Gabriela M. Baerlocher
- Department of Haematology; University Hospital and University of Berne; Berne Switzerland
| | - Katja Seipel
- Department of Clinical Research; University Hospital and University of Berne; Berne Switzerland
| | - Kurt Leibundgut
- Department of Paediatric Haemato-Oncology; University Hospital and University of Berne; Berne Switzerland
| | - Thomas Pabst
- Department of Medical Oncology; University Hospital and University of Berne; Berne Switzerland
| |
Collapse
|
56
|
Hatfield KJ, Reikvam H, Bruserud Ø. Identification of a subset of patients with acute myeloid leukemia characterized by long-termin vitroproliferation and altered cell cycle regulation of the leukemic cells. Expert Opin Ther Targets 2014; 18:1237-51. [DOI: 10.1517/14728222.2014.957671] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
57
|
Hu Y, Lv X, Wu Y, Xu J, Wang L, Chen W, Zhang W, Li J, Zhang S, Qiu H. Expression of costimulatory molecule B7-H3 and its prognostic implications in human acute leukemia. ACTA ACUST UNITED AC 2014; 20:187-95. [PMID: 25130683 DOI: 10.1179/1607845414y.0000000186] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVES This study focused on the expression pattern and clinical significance of B7-H3 expression in human acute leukemia. METHODS We systematically analyzed 134 patients with acute myeloid leukemia (101 cases) and acute lymphocytic leukemia (33 cases) by flow cytometry. RESULTS The frequency of B7-H3(+) cases was 44.8% in total. The B7-H3 expression rate differed from 0% to 74.8% in individual cases. The correlation between B7-H3 expression and traditional prognostic factors, such as age and gender, the white blood cell count was not confirmed. However, B7-H3 had a significant higher expression in CD34(+) cases and high risk karyotypes. CONCLUSIONS Owing to the expression of B7-H3 being statistically relevant in predicting disease progression and a shorter life survival, our results demonstrated that B7-H3 expression in acute leukemia predicts an unfavorable outcome.
Collapse
|
58
|
Rapin N, Porse BT. Oncogenic fusion proteins expressed in immature hematopoietic cells fail to recapitulate the transcriptional changes observed in human AML. Oncogenesis 2014; 3:e106. [PMID: 24932908 PMCID: PMC4150216 DOI: 10.1038/oncsis.2014.22] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 05/08/2014] [Accepted: 05/15/2014] [Indexed: 12/22/2022] Open
Abstract
Reciprocal chromosomal translocations are observed in one-third of acute myeloid leukemia (AML) cases. Targeting and understanding the effects of the resulting aberrant oncogenic fusion proteins may help developing drugs against specific leukemic subtypes, as demonstrated earlier by the use of ATRA in acute promyelocytic leukemia. Hematopoietic stem/progenitor (HSPCs) cells transduced with oncogenic fusion genes are regarded as promising in vitromodels of their corresponding AML subtypes. Here, we critically assessed the potential of such in vitro models using an integrative bioinformatics approach. Surprisingly, we found that the gene-expression profiles of CD34+ human HSPCs transformed with the potent oncogenic fusion proteins AML-ETO or MLL-AF9, only weakly resembled those derived from primary AML samples. Hence, our work raises concerns as to the relevance of the use of in vitro transduced cells to study the impact of transcriptional deregulation in human AML.
Collapse
Affiliation(s)
- N Rapin
- 1] The Finsen Laboratory, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark [2] Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark [3] Department of Biology, Faculty of Natural Sciences, The Bioinformatics Centre, University of Copenhagen, Copenhagen, Denmark [4] Danish Stem Cell Centre (DanStem) Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - B T Porse
- 1] The Finsen Laboratory, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark [2] Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark [3] Danish Stem Cell Centre (DanStem) Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
59
|
Cagnetta A, Adamia S, Acharya C, Patrone F, Miglino M, Nencioni A, Gobbi M, Cea M. Role of genotype-based approach in the clinical management of adult acute myeloid leukemia with normal cytogenetics. Leuk Res 2014; 38:649-59. [PMID: 24726781 DOI: 10.1016/j.leukres.2014.03.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 03/03/2014] [Accepted: 03/09/2014] [Indexed: 02/02/2023]
Abstract
Acute myeloid leukemia (AML) is the most common form of acute leukemia affecting adults. Although it is a complex disease driven by numerous genetic and epigenetic abnormalities, nearly 50% of patients exhibit a normal karyotype (CN-AML) with an intermediate cytogenetic risk. However, a widespread genomic analysis has recently shown the recurrence of genomic aberrations in this category (mutations of FLT3, CEBPA, NPM1, RUNX1, TET2, IDH1/2, DNMT3A, ASXL1, MLL and WT1) thus revealing its marked genomic heterogeneity. In this perspective, a global gene expression analysis of AML patients provides an independent prognostic marker to categorize each patient into clinic-pathologic subgroups based on its molecular genetic defects. Consistently such classification, taking into account the uniqueness of each AML patient, furnishes an individualized treatment approach leading a step closer to personalized medicine. Overall the genome-wide analysis of AML patients, by providing novel insights into biology of this tumor, furnishes accurate prognostic markers as well as useful tools for selecting the most appropriate treatment option. Moreover it provides novel therapeutic targets useful to enhance efficacy of the current anti-AML therapeutics. Here we describe the prognostic relevance of such new genetic data and discuss how this approach can be used to improve survival and treatment of AML patients.
Collapse
Affiliation(s)
- Antonia Cagnetta
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Department of Hematology and Oncology, I.R.C.C.S. A.O.U. San Martino-IST, Genoa, Italy.
| | - Sophia Adamia
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Chirag Acharya
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Franco Patrone
- Department of Hematology and Oncology, I.R.C.C.S. A.O.U. San Martino-IST, Genoa, Italy
| | - Maurizio Miglino
- Department of Hematology and Oncology, I.R.C.C.S. A.O.U. San Martino-IST, Genoa, Italy
| | - Alessio Nencioni
- Department of Hematology and Oncology, I.R.C.C.S. A.O.U. San Martino-IST, Genoa, Italy
| | - Marco Gobbi
- Department of Hematology and Oncology, I.R.C.C.S. A.O.U. San Martino-IST, Genoa, Italy
| | - Michele Cea
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Department of Hematology and Oncology, I.R.C.C.S. A.O.U. San Martino-IST, Genoa, Italy.
| |
Collapse
|
60
|
Prognostic significance of flow cytometric residual disease, dysregulated neutrophils/monocytes, and hematogones in adult acute myeloid leukemia in first remission. Int J Hematol 2014; 99:296-304. [PMID: 24481944 DOI: 10.1007/s12185-014-1525-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Revised: 01/15/2014] [Accepted: 01/15/2014] [Indexed: 10/25/2022]
Abstract
Fifty-one consecutive non-M3 acute myeloid leukemia (AML) patients who had achieved morphologic complete remission (mCR) after induction chemotherapy were enrolled in the present study. Three characteristics of bone marrow (BM) composition analyzed by flow cytometry were combined to determine the prognostic impact. A standardized panel of reagents was used to detect residual disease of aberrant myeloid progenitor cells (RD), identify neutrophils/monocytes with dysregulated immunophenotype (dysregulated neutro/mono) and quantify the appearance of CD34(+) B-progenitor-related cluster (hematogones) simultaneously in post-induction BM of adult AML patients. Patients who had detectable RD ≥0.2 % exhibited significantly lower median leukemia-free survival (LFS) than those who did not (13.5 vs. 48.0 months; P = 0.042). Dysregulated neutro/mono abnormalities assessed by this flow cytometric scoring system (FCSS ≥2) predicted shorter LFS (8.0 vs. 39.0 months; P = 0.008). While B-progenitor-related cluster size ≥5 % predicted improved outcome, with longer LFS (not reached vs. 13.5 months; P = 0.023) and better overall survival (not reached vs. 24.0 months; P = 0.027). The proposed RD/dysregulated neutro/mono/hematogones score showed a new risk groups with different LFS in the overall patients (P = 0.0006) as well as in the subgroup of intermediate cytogenetic risk (P = 0.001). The RD/dysregulated neutro/mono/hematogones score assessed by flow cytometry for adult AML in mCR may offer a rapid and practical risk assessment providing better refinement in risk-adapted management after induction chemotherapy.
Collapse
|
61
|
Rizzari C, Cazzaniga G, Coliva T, De Angelis C, Conter V. Predictive factors of relapse and survival in childhood acute myeloid leukemia: role of minimal residual disease. Expert Rev Anticancer Ther 2014; 11:1391-401. [DOI: 10.1586/era.11.37] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
62
|
|
63
|
|
64
|
A pipeline with multiplex reverse transcription polymerase chain reaction and microarray for screening of chromosomal translocations in leukemia. BIOMED RESEARCH INTERNATIONAL 2013; 2013:135086. [PMID: 24288660 PMCID: PMC3816023 DOI: 10.1155/2013/135086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 08/01/2013] [Accepted: 08/19/2013] [Indexed: 01/22/2023]
Abstract
Chromosome rearrangements and fusion genes present major portion of leukemogenesis and contribute to leukemic subtypes. It is practical and helpful to detect the fusion genes in clinic diagnosis of leukemia. Present application of reverse transcription polymerase chain reaction (RT-PCR) method to detect the fusion gene transcripts is effective, but time- and labor-consuming. To set up a simple and rapid system, we established a method that combined multiplex RT-PCR and microarray. We selected 15 clinically most frequently observed chromosomal rearrangements generating more than 50 fusion gene variants. Chimeric reverse primers and chimeric PCR primers containing both gene-specific and universal sequences were applied in the procedure of multiplex RT-PCR, and then the PCR products hybridized with a designed microarray. With this approach, among 200 clinic samples, 63 samples were detected to have gene rearrangements. All the detected fusion genes positive and negative were validated with RT-PCR and Sanger sequencing. Our data suggested that the RT-PCR-microarray pipeline could screen 15 partner gene pairs simultaneously at the same accuracy of the fusion gene detection with regular RT-PCR. The pipeline showed effectiveness in multiple fusion genes screening in clinic samples.
Collapse
|
65
|
Abstract
Recent advances in technological tools for massively parallel, high-throughput sequencing of DNA have enabled the comprehensive characterization of somatic mutations in a large number of tumour samples. In this Review, we describe recent cancer genomic studies that have assembled emerging views of the landscapes of somatic mutations through deep-sequencing analyses of the coding exomes and whole genomes in various cancer types. We discuss the comparative genomics of different cancers, including mutation rates and spectra, as well as the roles of environmental insults that influence these processes. We highlight the developing statistical approaches that are used to identify significantly mutated genes, and discuss the emerging biological and clinical insights from such analyses, as well as the future challenges of translating these genomic data into clinical impacts.
Collapse
Affiliation(s)
- Ian R Watson
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | |
Collapse
|
66
|
Freeman SD, Virgo P, Couzens S, Grimwade D, Russell N, Hills RK, Burnett AK. Prognostic relevance of treatment response measured by flow cytometric residual disease detection in older patients with acute myeloid leukemia. J Clin Oncol 2013; 31:4123-31. [PMID: 24062403 DOI: 10.1200/jco.2013.49.1753] [Citation(s) in RCA: 250] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
PURPOSE Older patients with acute myeloid leukemia (AML) have a high relapse rate after standard chemotherapy. We investigated whether measuring chemotherapy sensitivity by multiparameter flow cytometric minimal residual disease (MFC-MRD) detection has prognostic value in patients older than age 60 years or is simply a surrogate for known age-related risk factors. PATIENT AND METHODS Eight hundred ninety-two unselected patients treated intensively in the United Kingdom National Cancer Research Institute AML16 Trial were assessed prospectively for MFC-MRD during treatment. Eight hundred thirty-three patients had leukemia-associated immunophenotypes (LAIPs) identified by pretreatment screening. Four hundred twenty-seven patients entered complete remission (CR) after one or two courses (designated C1 and C2, respectively) and were MFC-MRD assessable by LAIP detection in CR bone marrow for at least one of these time points. MRD positivity was defined as residual disease detectable by LAIP. RESULTS MFC-MRD negativity, which was achieved in 51% of patients after C1 (n = 286) and 64% of patients after C2 (n = 279), conferred significantly better 3-year survival from CR (C1: 42% v 26% in MRD-positive patients, P < .001; C2: 38% v 18%, respectively; P < .001) and reduced relapse (C1: 71% v 83% in MRD-positive patients, P < .001; C2: 79% v 91%, respectively; P < .001), with higher risk of early relapse in MRD-positive patients (median time to relapse, 8.5 v 17.1 months, respectively). In multivariable analysis, MRD status at the post-C1 time point independently predicted survival, identifying a subgroup of intermediate-risk patients with particularly poor outcome. However, survival benefit from gemtuzumab ozogamicin was not associated with MFC-MRD chemotherapy sensitivity. CONCLUSION Early assessment of treatment response using flow cytometry provides powerful independent prognostic information in older adults with AML, lending support to the incorporation of MRD detection to refine risk stratification and inform clinical trial design in this challenging group of patients.
Collapse
Affiliation(s)
- Sylvie D Freeman
- Sylvie D. Freeman, University of Birmingham and University Hospitals Birmingham National Health Service (NHS) Trust, Birmingham; Paul Virgo, North Bristol NHS Trust, Bristol; Steve Couzens, University Hospital of Wales; Robert K. Hills and Alan K. Burnett, Cardiff University, Heath Park, Cardiff; David Grimwade, King's College London School of Medicine and Guy's and St Thomas' NHS Foundation Trust, London; and Nigel Russell, Nottingham University Hospital NHS Trust, Nottingham, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
67
|
Terwijn M, van Putten WLJ, Kelder A, van der Velden VHJ, Brooimans RA, Pabst T, Maertens J, Boeckx N, de Greef GE, Valk PJM, Preijers FWMB, Huijgens PC, Dräger AM, Schanz U, Jongen-Lavrecic M, Biemond BJ, Passweg JR, van Gelder M, Wijermans P, Graux C, Bargetzi M, Legdeur MC, Kuball J, de Weerdt O, Chalandon Y, Hess U, Verdonck LF, Gratama JW, Oussoren YJM, Scholten WJ, Slomp J, Snel AN, Vekemans MC, Löwenberg B, Ossenkoppele GJ, Schuurhuis GJ. High prognostic impact of flow cytometric minimal residual disease detection in acute myeloid leukemia: data from the HOVON/SAKK AML 42A study. J Clin Oncol 2013; 31:3889-97. [PMID: 24062400 DOI: 10.1200/jco.2012.45.9628] [Citation(s) in RCA: 341] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Half the patients with acute myeloid leukemia (AML) who achieve complete remission (CR), ultimately relapse. Residual treatment-surviving leukemia is considered responsible for the outgrowth of AML. In many retrospective studies, detection of minimal residual disease (MRD) has been shown to enable identification of these poor-outcome patients by showing its independent prognostic impact. Most studies focus on molecular markers or analyze data in retrospect. This study establishes the value of immunophenotypically assessed MRD in the context of a multicenter clinical trial in adult AML with sample collection and analysis performed in a few specialized centers. PATIENTS AND METHODS In adults (younger than age 60 years) with AML enrolled onto the Dutch-Belgian Hemato-Oncology Cooperative Group/Swiss Group for Clinical Cancer Research Acute Myeloid Leukemia 42A study, MRD was evaluated in bone marrow samples in CR (164 after induction cycle 1, 183 after cycle 2, 124 after consolidation therapy). RESULTS After all courses of therapy, low MRD values distinguished patients with relatively favorable outcome from those with high relapse rate and adverse relapse-free and overall survival. In the whole patient group and in the subgroup with intermediate-risk cytogenetics, MRD was an independent prognostic factor. Multivariate analysis after cycle 2, when decisions about consolidation treatment have to be made, confirmed that high MRD values (> 0.1% of WBC) were associated with a higher risk of relapse after adjustment for consolidation treatment time-dependent covariate risk score and early or later CR. CONCLUSION In future treatment studies, risk stratification should be based not only on risk estimation assessed at diagnosis but also on MRD as a therapy-dependent prognostic factor.
Collapse
Affiliation(s)
- Monique Terwijn
- Monique Terwijn, Angèle Kelder, Peter C. Huijgens, Angelika M. Dräger, Yvonne J.M. Oussoren, Willemijn J. Scholten, Alexander N. Snel, Gert J. Ossenkoppele, and Gerrit J. Schuurhuis, VU University Medical Centre; Bart J. Biemond, Academic Medical Centre, Amsterdam; Wim L.J. van Putten, Vincent H.J. van der Velden, Georgine E. de Greef, Peter J.M. Valk, Mojca Jongen-Lavrecic, and Bob Löwenberg, Erasmus University Medical Centre; Rik A. Brooimans, Jan W. Gratama, Erasmus University Medical Centre/Daniel den Hoed Cancer Centre, Rotterdam; Frank W.M.B. Preijers, Radboud University Nijmegen Medical Center, Nijmegen; Michel van Gelder, University Medical Centre, Maastricht; Pierre Wijermans, Haga Hospital, The Hague; Marie-Cecile Legdeur and Jennita Slomp, Medisch Spectrum Twente, Enschede; Jurgen Kuball, University Medical Center Utrecht; Okke de Weerdt, St. Antonius Hospital, Nieuwegein; Leo F. Verdonck, Isala Clinics, Zwolle, the Netherlands; Thomas Pabst, Bern University Hospital, Bern; Urs Schanz, University Hospital, Zürich; Jakob R. Passweg, Basel University Hospital, Basel; Mario Bargetzi, Kantonspital, Aarau; Yves Chalandon, University Hospital of Geneva, Geneva; Urs Hess, Kantonsspital, St. Gallen, Switzerland; Johan Maertens, University Hospital Gasthuisberg; Nancy Boeckx, University Hospitals Leuven and Katholieke Universiteit Leuven, Leuven; Carlos Graux, Cliniques Universitaires-Université Catholique de Louvain, Mont-Godinne, Yvoir; and Marie-Christiane Vekemans, St. Luc Hospital, Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Wang Y, Zhang L, Chen WL, Wang JH, Li N, Li JM, Mi JQ, Zhang WN, Li Y, Wu SF, Jin J, Wang YG, Huang H, Chen Z, Chen SJ, Tang H. Rapid diagnosis and prognosis of de novo acute myeloid leukemia by serum metabonomic analysis. J Proteome Res 2013; 12:4393-401. [PMID: 23998518 DOI: 10.1021/pr400403p] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Acute myeloid leukemia (AML) is a life-threatening hematological disease. Novel diagnostic and prognostic markers will be essential for new therapeutics and for significantly improving the disease prognosis. To characterize the metabolic features associated with AML and search for potential diagnostic and prognostic methods, here we analyzed the phenotypic characteristics of serum metabolite composition (metabonome) in a cohort of 183 patients with de novo acute myeloid leukemia together with 232 age- and gender-matched healthy controls using (1)H NMR spectroscopy in conjunction with multivariate data analysis. We observed significant serum metabonomic differences between AML patients and healthy controls and between AML patients with favorable and intermediate cytogenetic risks. Such differences were highlighted by systems differentiations in multiple metabolic pathways including glycolysis/gluconeogenesis, TCA cycle, biosynthesis of proteins and lipoproteins, and metabolism of fatty acids and cell membrane components, especially choline and its phosphorylated derivatives. This demonstrated the NMR-based metabonomics as a rapid and less invasive method for potential AML diagnosis and prognosis. The serum metabolic phenotypes observed here indicated that integration of metabonomics with other techniques will be useful for better understanding the biochemistry of pathogenesis and progression of leukemia.
Collapse
Affiliation(s)
- Yihuang Wang
- State Key Laboratory of Medical Genomics, Institute of Health Sciences, Shanghai Institutes for Biological Sciences and Graduate School, Chinese Academy of Sciences and Shanghai Institute of Hematology, Rui Jin Hospital, affiliated to Shanghai Jiao Tong University School of Medicine , Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
69
|
FLT3-ITD and MLL-PTD influence the expression of MDR-1, MRP-1, and BCRP mRNA but not LRP mRNA assessed with RQ-PCR method in adult acute myeloid leukemia. Ann Hematol 2013; 93:577-93. [PMID: 24030729 DOI: 10.1007/s00277-013-1898-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 08/30/2013] [Indexed: 12/20/2022]
Abstract
Fms-like tyrosine kinase 3-internal tandem duplication (FLT3-ITD) and mixed-lineage leukemia gene-partial tandem duplication (MLL-PTD) are aberrations associated with leukemia which indicate unsatisfactory prognosis. Downstream regulatory targets of FLT3-ITD and MLL-PTD are not well defined. We have analyzed the expression of MDR-1, multidrug resistant protein-1 (MRP-1), breast cancer resistance protein (BCRP), and lung resistance protein (LRP) messenger RNA (mRNA) in relation to the mutational status of FLT3-ITD and MLL-PTD in 185 acute myeloid leukemia (AML) adult patients. The real-time quantitative polymerase chain reaction method was performed to assess the expression of the MDR-1, MRP-1, BCRP, and LRP mRNA, and the results were presented as coefficients calculated using an intermediate method according to Pfaffl's rule. Significantly higher expressions of MDR-1 mRNA were found in patients who did not harbor FLT3-ITD (0.20 vs. 0.05; p = 0.0001) and MRP-1 mRNA in patients with this mutation (0.96 vs. 0.70; p = 0.002) and of BCRP mRNA in patients with MLL-PTD (0.61 vs. 0.38; p = 0.03). In univariate analysis, the high expression of MDR-1 mRNA (≥0.1317) negatively influenced the outcome of induction therapy (p = 0.05), whereas the high expression of BCRP mRNA (≥1.1487) was associated with a high relapse rate (RR) (p = 0.013). We found that the high expression of MDR-1 (≥0.1317), MRP-1 (≥0.8409), and BCRP mRNA (≥1.1487) significantly influenced disease-free survival (DFS; p = 0.059, 0.032, and 0.009, respectively) and overall survival (0.048, 0.014, and 0.059, respectively). Moreover, a high expression of BCRP mRNA (≥1.1487) proved to be an independent prognostic factor for RR (p = 0.01) and DFS (p = 0.002) in multivariate analysis. The significant correlation between the expression of MDR-1, MRP-1, and BCRP mRNA and FLT3-ITD or MLL-PTD in AML patients requires further investigation.
Collapse
|
70
|
Hoyos M, Nomdedeu JF, Esteve J, Duarte R, Ribera JM, Llorente A, Escoda L, Bueno J, Tormo M, Gallardo D, de Llano MPQ, Martí JM, Aventín A, Mangues R, Brunet S, Sierra J. Core binding factor acute myeloid leukemia: the impact of age, leukocyte count, molecular findings, and minimal residual disease. Eur J Haematol 2013; 91:209-218. [PMID: 23646898 DOI: 10.1111/ejh.12130] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2013] [Indexed: 12/31/2022]
Abstract
PURPOSE Most patients with acute myeloid leukemia (AML) and genetic rearrangements involving the core binding factor (CBF) have favorable prognosis. In contrast, a minority of them still have a high risk of leukemia recurrence. This study investigated the adverse features of CBF AML that could justify investigational therapeutic approaches. PATIENTS AND METHODS One hundred and fifty patients (median age 42 yr, range 16-69) with CBF AML (RUNX1-RUNX1T1 n = 74; CBFB-MYH11 n = 76) were prospectively enrolled into two consecutive CETLAM protocols at 19 Spanish institutions. Main clinic and biologic parameters were analyzed in the whole series. In non-selected cases with available DNA samples, the impact of molecular characterization and minimal residual disease (MRD) was also studied. RESULTS Overall, complete remission (CR) rate was 89% (94% in ≤50 yr old and 72% in >50 yr, P = 0.002). At 5 yr, cumulative incidence of relapse (CIR) was 26 ± 1%, disease-free survival (DFS) 62 ± 6%, and overall survival (OS) 66 ± 4%. In multivariate analyses, leukocyte count above 20 × 10(9) /L, BAALC over-expression, and high copy numbers of RUNX1-RUNXT1 or CBFB-MYH11 after induction chemotherapy (CT) led to increased relapse rate. Regarding OS, age >50 yr, leukocyte count above 20 × 10(9) /L, and increased MN1 expression were adverse features. CONCLUSION Age, leukocyte counts, BAALC, and MN1 gene expressions as well as high copy numbers of RUNX1-RUNXT1 or CBFB-MYH11 after induction chemotherapy are useful tools to predict the outcome and should be considered for risk-adapted therapy.
Collapse
Affiliation(s)
- Montserrat Hoyos
- Spanish CETLAM Group Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona Spanish Cancer Network (RTICC), Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain; University of Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Kantarjian HM, Martinelli G, Jabbour EJ, Quintás-Cardama A, Ando K, Bay JO, Wei A, Gröpper S, Papayannidis C, Owen K, Pike L, Schmitt N, Stockman PK, Giagounidis A. Stage I of a phase 2 study assessing the efficacy, safety, and tolerability of barasertib (AZD1152) versus low-dose cytosine arabinoside in elderly patients with acute myeloid leukemia. Cancer 2013; 119:2611-9. [PMID: 23605952 PMCID: PMC4132839 DOI: 10.1002/cncr.28113] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 03/07/2013] [Accepted: 03/12/2013] [Indexed: 11/05/2022]
Abstract
BACKGROUND In this phase 2 study, the authors evaluated the efficacy, safety, and tolerability of the Aurora B kinase inhibitor barasertib compared with low-dose cytosine arabinoside (LDAC) in patients aged ≥ 60 years with acute myeloid leukemia (AML). METHODS Patients were randomized 2:1 to receive either open-label barasertib 1200 mg (as a 7-day intravenous infusion) or LDAC 20 mg (subcutaneously twice daily for 10 days) in 28-day cycles. The primary endpoint was the objective complete response rate (OCRR) (complete responses [CR] plus confirmed CRs with incomplete recovery of neutrophils or platelets [CRi] according to Cheson criteria [also requiring reconfirmation of CRi ≥21 days after the first appearance and associated with partial recovery of platelets and neutrophils]). Secondary endpoints included overall survival (OS) and safety. RESULTS In total, 74 patients (barasertib, n = 48; LDAC, n = 26) completed ≥1 cycle of treatment. A significant improvement in the OCRR was observed with barasertib (35.4% vs 11.5%; difference, 23.9%; 95% confidence interval, 2.7%-39.9%; P < .05). Although the study was not formally sized to compare OS data, the median OS with barasertib was 8.2 months versus 4.5 months with LDAC (hazard ratio, 0.88; 95% confidence interval, 0.49-1.58; P = .663). Stomatitis and febrile neutropenia were the most common adverse events with barasertib versus LDAC (71% vs 15% and 67% vs 19%, respectively). CONCLUSIONS Barasertib produced a significant improvement in the OCRR versus LDAC and had a more toxic but manageable safety profile, consistent with previous studies.
Collapse
|
72
|
Upegui RI, Betes VM, Solé F, López R. [Spontaneous remission of bone marrow aplasia associated with chromosome 8 trisomy, with subsequent evolution to acute monoblastic leukemia and recurrence of the clonal abnormality]. Med Clin (Barc) 2013; 141:42-3. [PMID: 23276618 DOI: 10.1016/j.medcli.2012.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Revised: 10/11/2012] [Accepted: 10/18/2012] [Indexed: 11/29/2022]
MESH Headings
- Adult
- Anemia, Aplastic/diagnosis
- Anemia, Aplastic/etiology
- Anemia, Aplastic/genetics
- Anemia, Aplastic/pathology
- Bone Marrow/pathology
- Chromosomes, Human, Pair 8/genetics
- Clone Cells/ultrastructure
- Fatal Outcome
- Fever of Unknown Origin/etiology
- Humans
- In Situ Hybridization, Fluorescence
- Karyotyping
- Leukemia, Monocytic, Acute/genetics
- Leukemia, Monocytic, Acute/pathology
- Male
- Neoplastic Stem Cells/ultrastructure
- Recurrence
- Remission, Spontaneous
- Trisomy
Collapse
|
73
|
Shah A, Andersson TML, Rachet B, Björkholm M, Lambert PC. Survival and cure of acute myeloid leukaemia in England, 1971-2006: a population-based study. Br J Haematol 2013; 162:509-16. [PMID: 23786647 DOI: 10.1111/bjh.12425] [Citation(s) in RCA: 149] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 05/09/2013] [Indexed: 12/27/2022]
Abstract
The 5-year relative survival of adults diagnosed with acute myeloid leukaemia (AML) was less than 10% during the 1970s and 1980s in England. This population-based study estimated the 5-year relative survival and 'cure' for 48 380 adult patients diagnosed with AML in England during 1971-2006. Relative survival and cure mixture models were used to produce estimates of 5-year relative survival and the percentage 'cured'. 'Cure' was defined as the proportion of a group of survivors for whom there is no excess mortality compared with the general population. The 5-year relative survival and the percentage 'cured' increased for patients aged under 70 years at diagnosis during 1971-2006, but advancing age was associated with poorer outcome. During the study period a dramatic increase in 5-year relative survival occurred in those aged 15-24 years, from 7% to 53%. The percentage 'cured' was less than 10% for all ages in 1975, but increased to 45% for those aged 15-24 years in 2000. Cure could not be estimated for patients over 70 years, because survival was consistently low (<5%). The long-term outcome of patients with AML has improved substantially, particularly in younger patients. The potential exists for further increasing levels of 'cure'.
Collapse
Affiliation(s)
- Anjali Shah
- Cancer Research UK Cancer Survival Group, London School of Hygiene and Tropical Medicine, London, UK.
| | | | | | | | | |
Collapse
|
74
|
Kantarjian HM, Sekeres MA, Ribrag V, Rousselot P, Garcia-Manero G, Jabbour EJ, Owen K, Stockman PK, Oliver SD. Phase I study assessing the safety and tolerability of barasertib (AZD1152) with low-dose cytosine arabinoside in elderly patients with AML. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2013; 13:559-67. [PMID: 23763917 DOI: 10.1016/j.clml.2013.03.019] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 03/06/2013] [Accepted: 03/27/2013] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Barasertib is the pro-drug of barasertib-hydroxy-quinazoline pyrazole anilide, a selective Aurora B kinase inhibitor that has demonstrated preliminary anti-AML activity in the clinical setting. PATIENTS AND METHODS This Phase I dose-escalation study evaluated the safety and tolerability of barasertib, combined with LDAC, in patients aged 60 years or older with de novo or secondary AML. Barasertib (7-day continuous intravenous infusion) plus LDAC 20 mg (subcutaneous injection twice daily for 10 days) was administered in 28-day cycles. The MTD was defined as the highest dose at which ≤ 1 patient within a cohort of 6 experienced a dose-limiting toxicity (DLT) (clinically significant adverse event [AE] or laboratory abnormality considered related to barasertib). The MTD cohort was expanded to 12 patients. RESULTS Twenty-two patients (median age, 71 years) received ≥ 1 treatment cycle (n = 6, 800 mg; n = 13, 1000 mg; n = 3, 1200 mg). DLTs were reported in 2 patients (both, National Cancer Institute Common Terminology Criteria for Adverse Events grade 3 stomatitis/mucositis; 1200 mg cohort). The most common AEs were infection (73%), febrile neutropenia (59%), nausea (50%), and diarrhea (46%). Barasertib plus LDAC resulted in an overall response rate (International Working Group criteria) of 45% (n = 10/22; according to investigator opinion). CONCLUSION The MTD of 1000 mg barasertib in combination with LDAC in older patients with AML was associated with acceptable tolerability and preliminary anti-AML activity.
Collapse
|
75
|
Costa ARDS, Vasudevan A, Krepischi A, Rosenberg C, Chauffaille MDLLF. Single-nucleotide polymorphism-array improves detection rate of genomic alterations in core-binding factor leukemia. Med Oncol 2013; 30:579. [PMID: 23636907 DOI: 10.1007/s12032-013-0579-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 04/10/2013] [Indexed: 11/30/2022]
Abstract
Acute myeloid leukemia (AML) is a group of clonal diseases, resulting from two classes of mutation. Investigation for additional abnormalities associated with a well-recognized subtype, core-binding factor AML (CBF-AML) can provide further understanding and discrimination to this special group of leukemia. In order to better define genetic alterations in CBF-AML and identify possible cooperating lesions, a single-nucleotide polymorphism-array (SNP-array) analysis was performed, combined to KIT mutation screening, in a set of cases. Validation of SNP-array results was done by array comparative genomic hybridization and FISH. Fifteen cases were analyzed. Three cases had microscopic lesions better delineated by arrays. One case had +22 not identified by arrays. Submicroscopic abnormalities were mostly non-recurrent between samples. Of relevance, four regions were more frequently affected: 4q28, 9p11, 16q22.1, and 16q23. One case had an uncovered unbalanced inv(16) due to submicroscopic deletion of 5´MYH11 and 3´CBFB. Telomeric and large copy number neutral loss of heterozygosity (CNN-LOH) regions (>25 Mb), likely representing uniparental disomy, were detected in four out of fifteen cases. Only three cases had mutation on KIT gene, enhancing the role of abnormalities by SNP-array as presumptive cooperating alterations. Molecular karyotyping can add valuable information to metaphase karyotype analysis, emerging as an important tool to uncover and characterize microscopic, submicroscopic genomic alterations, and CNN-LOH events in the search for cooperating lesions.
Collapse
Affiliation(s)
- Ana Rosa da Silveira Costa
- Division of Hematology and Hemotherapy, UNIFESP/Escola Paulista de Medicina, São Paulo, SP, CEP: 04037-002, Brazil.
| | | | | | | | | |
Collapse
|
76
|
Shah A, Seedhouse C. Frequency of TP53 Mutations and its Impact on Drug Sensitivity in Acute Myeloid Leukemia? Indian J Clin Biochem 2013; 27:121-6. [PMID: 23543587 DOI: 10.1007/s12291-012-0203-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Accepted: 03/03/2012] [Indexed: 11/29/2022]
Abstract
The purpose of this study is to find out the frequency of TP53 mutations in acute myeloid leukemia (AML) patients and correlate sensitivity of drug response with TP53 mutations. In AML more than 90 % of cases comprise of wild type TP53. 94.2 % of TP53 mutations are found within exon 5-8 of which 73 % are point mutations. TP53 mutations were analysed with high resolution melting curve analysis. We analysed 106 AML samples of which we found nine mutations which represents 8.5 % mutation rate and found one rare SNP. The effect of TP53 mutations were studied on the chemosensitivity of two new drugs AZD115 and RHPS4, an Aurora Kinase B inhibitor and Telomerase inhibitor respectively. Four mutations were found out of 17 for RHPS4 stating significant (p = 0.002) increase in sensitivity and no mutation found in AZD1152 database, but need more study to get definite conclusion.
Collapse
Affiliation(s)
- Ankur Shah
- Department of Molecular Medical Sciences, Queens Medical Centre, University of Nottingham, Nottingham, NG7 2UH UK
| | | |
Collapse
|
77
|
Kampen KR, Ter Elst A, de Bont ESJM. Vascular endothelial growth factor signaling in acute myeloid leukemia. Cell Mol Life Sci 2013; 70:1307-17. [PMID: 22833169 PMCID: PMC11113417 DOI: 10.1007/s00018-012-1085-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Revised: 06/19/2012] [Accepted: 07/09/2012] [Indexed: 01/08/2023]
Abstract
This review is designed to provide an overview of the current literature concerning vascular endothelial growth factor signaling (VEGF) in acute myeloid leukemia (AML). Aberrant VEGF signaling operates in the bone marrow of AML patients and is related to a poor prognosis. The altered signaling pathway demonstrated to interfere in several autocrine and paracrine signaling pathways. VEGF signaling promotes autocrine AML blast cell proliferation, survival, and chemotherapy resistance. In addition, VEGF signaling can mediate paracrine vascular endothelial cell-controlled angiogenesis in AML. Both effects presumably explain the association of high VEGF levels and poor therapeutic outcome. More recently, researches focusing on bone marrow stem cell niches demonstrate a role for VEGF signaling in the preservation of several cell types within these niches. The bone marrow niches are proposed to be a protective microenvironment for AML cells that could be responsible for relapses in AML patients. This implies the need of sophisticated VEGF-targeted therapeutics in AML therapy strategies. This review highlights our current understanding of aberrant VEGF signaling in AML, appoints the interference of VEGF signaling in the AML-associated microenvironment, and reflects the novelty of current VEGF-targeted therapeutics used in clinical trails for the treatment of AML.
Collapse
Affiliation(s)
- Kim R Kampen
- Division of Pediatric Oncology/Hematology, Department of Pediatrics, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, PO Box 30.001, 9700 RB, Groningen, The Netherlands.
| | | | | |
Collapse
|
78
|
Mupo A, Celani L, Dovey O, Cooper JL, Grove C, Rad R, Sportoletti P, Falini B, Bradley A, Vassiliou GS. A powerful molecular synergy between mutant Nucleophosmin and Flt3-ITD drives acute myeloid leukemia in mice. Leukemia 2013; 27:1917-20. [PMID: 23478666 PMCID: PMC3768110 DOI: 10.1038/leu.2013.77] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
79
|
Fasan A, Alpermann T, Haferlach C, Grossmann V, Roller A, Kohlmann A, Eder C, Kern W, Haferlach T, Schnittger S. Frequency and prognostic impact of CEBPA proximal, distal and core promoter methylation in normal karyotype AML: a study on 623 cases. PLoS One 2013; 8:e54365. [PMID: 23383300 PMCID: PMC3562230 DOI: 10.1371/journal.pone.0054365] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 12/11/2012] [Indexed: 11/26/2022] Open
Abstract
The clinical impact of aberrant CEBPA promoter methylation (PM) in AML is controversially discussed. The aim of this study was to clarify the significance of aberrant CEBPA PM with regard to clinical features in a cohort of 623 cytogenetically normal (CN) de novo AML. 555 cases had wild-type CEBPA, 68 cases harbored CEBPA mutations. The distal promoter was methylated in 238/623 cases (38.2%), the core promoter in 8 of 326 cases (2.5%), whereas proximal PM was never detected. CEBPA PM and CEBPA mutations were mutually exclusive. CEBPA distal PM positive cases were characterized by reduced CEBPA mRNA expression levels and elevated white blood cell counts. CEBPA distal PM was less frequent in patients with mutations in FLT3, NPM1 and TET2 and more frequent in cases with RUNX1 and IDH2R140 mutations. Overall, no association of methylation to prognosis was seen. However CEBPA distal PM was associated with inferior outcome in cases with low FLT3-ITD ratio or TET2 mutations. A distinct gene expression profile of CEBPA distal PM positive cases compared to CEBPA mutated and CEBPA distal PM negative cases was observed. In conclusion, the presence of aberrant CEBPA PM is associated with distinct biological features but impact on outcome is weak.
Collapse
|
80
|
Djunic I, Virijevic M, Novkovic A, Djurasinovic V, Colovic N, Vidovic A, Suvajdzic-Vukovic N, Tomin D. Pretreatment risk factors and importance of comorbidity for overall survival, complete remission, and early death in patients with acute myeloid leukemia. ACTA ACUST UNITED AC 2012; 17:53-8. [PMID: 22664041 DOI: 10.1179/102453312x13221316477651] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
The objective of this single-center study was to determine the pretreatment risk factors and influence of comorbidity on outcome in patients with acute myeloid leukemia (AML). The research involved 145 patients with AML during a 58-month follow-up period. The results suggest that the most significant predictor of poor overall survival (OS) is an adverse karyotype (P = 0.007), while for poor rate of complete remission (CR) it is age ≥55 years, and for early death the most significant predictor is comorbidity, as scored by the Hematopoetic Cell Transplantation Comorbidity Index (HCT-CI), P = 0.001. When we divided the patients into two groups: aged ≥55 years and aged <55 years, these predictors differed. In the group aged ≥55 years the most significant predictor of OS (P = 0.013) and for early death (P = 0.003) was HCT-CI (P = 0.013), while in the younger group it was karyotype (P < 0.001). The most significant predictor of CR in the elderly was increased serum lactate dehydrogenase (LDH) level (P = 0.045). In the younger patients, the most significant predictor of CR was leukocytosis (P = 0.001) and for early death it was infection as the comorbidity (P = 0.007). We point out the importance of comorbidity for OS and early death, as well as the impact of infection in patients with AML.
Collapse
Affiliation(s)
- Irena Djunic
- Clinic for Hematology, Clinical Center of Serbia, Belgrade, Serbia.
| | | | | | | | | | | | | | | |
Collapse
|
81
|
Grimwade D. The changing paradigm of prognostic factors in acute myeloid leukaemia. Best Pract Res Clin Haematol 2012. [DOI: 10.1016/j.beha.2012.10.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
82
|
Lin A, Othus M, McQuary A, Chi M, Estey E. Influence of obesity on efficacy and toxicity of induction chemotherapy in patients with newly diagnosed acute myeloid leukemia. Leuk Lymphoma 2012; 54:541-6. [PMID: 22852586 DOI: 10.3109/10428194.2012.717278] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Increased body mass index (BMI) is associated with increased risk of treatment-related complications and inferior overall survival in patients with acute myeloid leukemia (AML). We retrospectively evaluated the association between percentage of ideal body weight (IBW) and complete remission (CR) among 63 newly diagnosed, previously untreated patients with AML. The median percentage of ideal body weight was 121% (range 86-246%). Thirty-three percent of patients were obese (≥ 130% IBW). In multivariate analysis, obesity was not associated with CR (odds ratio [OR] = 0.97, p = 0.88), overall survival (hazard ratio = 0.48, p = 0.52), platelet recovery by 30 days (OR = 1.14, p = 0.52) or neutrophil recovery by 30 days (OR = 1.12, p = 0.60). Obesity was also not associated with any differences in non-hematologic toxicity. CR rates were not significantly different comparing patients not dose-adjusted to patients with obesity-related adjustments (CR = 86% vs. 67%, p = 0.55). Empiric dose reductions based on obesity did not result in significantly different CR rates.
Collapse
Affiliation(s)
- Andrew Lin
- University of Washington Medical Center, Seattle, WA, USA.
| | | | | | | | | |
Collapse
|
83
|
Abstract
The karyotype is so far the most important prognostic parameter in acute myeloid leukemia (AML). Molecular mutations have been analyzed to subdivide AML with normal karyotype into prognostic subsets. The aim of this study was to develop a prognostic model for the entire AML cohort solely based on molecular markers. One thousand patients with cytogenetic data were investigated for the following molecular alterations: PML-RARA, RUNX1-RUNX1T1, CBFB-MYH11, FLT3-ITD, and MLL-PTD, as well as mutations in NPM1, CEPBA, RUNX1, ASXL1, and TP53. Clinical data were available in 841 patients. Based on Cox regression and Kaplan-Meier analyses, 5 distinct prognostic subgroups were identified: (1) very favorable: PML-RARA rearrangement (n = 29) or CEPBA double mutations (n = 42; overall survival [OS] at 3 years: 82.9%); (2) favorable: RUNX1-RUNX1T1 (n = 35), CBFB-MYH11 (n = 31), or NPM1 mutation without FLT3-ITD (n = 186; OS at 3 years: 62.6%); (3) intermediate: none of the mutations leading to assignment into groups 1, 2, 4, or 5 (n = 235; OS at 3 years: 44.2%); (4) unfavorable: MLL-PTD and/or RUNX1 mutation and/or ASXL1 mutation (n = 203; OS at 3 years: 21.9%); and (5) very unfavorable: TP53 mutation (n = 80; OS at 3 years: 0%; P < .001). This comprehensive molecular characterization provides a more powerful model for prognostication than cytogenetics.
Collapse
|
84
|
The superiority of haploidentical related stem cell transplantation over chemotherapy alone as postremission treatment for patients with intermediate- or high-risk acute myeloid leukemia in first complete remission. Blood 2012; 119:5584-90. [PMID: 22535659 DOI: 10.1182/blood-2011-11-389809] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Abstract
We report the results of a prospective, patient self-selected study evaluating whether haploidentical related donor stem cell transplantation (HRD-HSCT) is superior to chemotherapy alone as postremission treatment for patients with intermediate- or high-risk acute myeloid leukemia (AML) in first complete remission (CR1). Among totally 419 newly diagnosed AML patients, 132 patients with intermediate- and high-risk cytogenetics achieved CR1 and received chemotherapy alone (n = 74) or HSCT (n = 58) as postremission treatment. The cumulative incidence of relapse at 4 years was 37.5% ± 4.5%. Overall survival (OS) and disease-free survival (DFS) at 4 years were 64.5% ± 5.1% and 55.6% ± 5.0%, respectively. The cumulative incident of relapse for the HRD-HSCT group was significantly lower than that for the chemotherapy-alone group (12.0% ± 4.6% vs 57.8% ± 6.2%, respectively; P < .0001). HRD-HSCT resulted in superior survival compared with chemotherapy alone (4-year DFS, 73.1% ± 7.1% vs 44.2% ± 6.2%, respectively; P < .0001; 4-year OS, 77.5% ± 7.1% vs 54.7% ± 6.3%, respectively; P = .001). Multivariate analysis revealed postremission treatment (HRD-HSCT vs chemotherapy) and high WBC counts at diagnosis as independent risk factors affecting relapse, DFS, and OS. Our results suggest that HRD-HSCT is superior to chemotherapy alone as postremission treatment for AML.
Collapse
|
85
|
Inaba H, Surprise HC, Pounds S, Cao X, Howard SC, Ringwald-Smith K, Buaboonnam J, Dahl G, Bowman WP, Taub JW, Campana D, Pui CH, Ribeiro RC, Rubnitz JE. Effect of body mass index on the outcome of children with acute myeloid leukemia. Cancer 2012; 118:5989-96. [PMID: 22648558 DOI: 10.1002/cncr.27640] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2012] [Revised: 04/03/2012] [Accepted: 04/10/2012] [Indexed: 11/12/2022]
Abstract
BACKGROUND The effect of body mass index (BMI) on the treatment outcomes of children with acute myeloid leukemia (AML) is unclear and needs further evaluation. METHODS Children with AML (n = 314) who were enrolled in 4 consecutive St. Jude protocols were grouped according to BMI (underweight, <5th percentile; healthy weight, 5th to 85th percentile; and overweight/obese, ≥ 85th percentile). RESULTS Twenty-five patients (8%) were underweight, 86 patients (27.4%) were overweight/obese, and 203 patients (64.6%) had healthy weight. The 5-year overall survival rate of overweight/obese patients (46.5% ± 7.3%) was lower than the rate of patients with healthy weight (67.1% ± 4.3%; P < .001); underweight patients also tended to have lower survival rates (50.6% ± 10.7%; P = .18). In a multivariable analysis that was adjusted for age, leukocyte count, French-American-British classification, and study protocols, patients with healthy weight had the best survival rate among the 3 groups (P = .01). When BMI was considered as continuous variable, patients with lower or higher BMI percentiles had worse survival (P = .03). There was no difference in the occurrence of induction failure or relapse among BMI groups, although underweight and overweight/obese patients had a significantly higher cumulative incidence of treatment-related mortality, especially because of infection (P = .009). CONCLUSIONS An unhealthy BMI was associated with worse survival and more treatment-related mortality in children with AML. Meticulous supportive care with nutritional support and education, infection prophylaxis, and detailed laboratory and physical examination is required for these patients. These measures, together with pharmacokinetics-guided chemotherapy dosing, may improve outcome.
Collapse
Affiliation(s)
- Hiroto Inaba
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105-2794, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Cornelissen JJ, Breems D, van Putten WLJ, Gratwohl AA, Passweg JR, Pabst T, Maertens J, Beverloo HB, van Marwijk Kooy M, Wijermans PW, Biemond BJ, Vellenga E, Verdonck LF, Ossenkoppele GJ, Löwenberg B. Comparative analysis of the value of allogeneic hematopoietic stem-cell transplantation in acute myeloid leukemia with monosomal karyotype versus other cytogenetic risk categories. J Clin Oncol 2012; 30:2140-6. [PMID: 22564995 DOI: 10.1200/jco.2011.39.6499] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To determine to what extent allogeneic hematopoietic stem-cell transplantation (alloHSCT) quantitatively reduces relapse in acute myeloid leukemia with monosomal karyotype (MK-AML) compared with alternative postremission therapy and how it compares with other cytogenetic subcategories. PATIENTS AND METHODS Of 2,560 patients (younger than age 61 years) without core-binding factor abnormalities including 305 patients with MK-AML receiving first-line induction treatment, 1,975 patients (77%) achieved remission, and 1,588 received consolidation in the first complete remission (CR1) after two induction cycles. Consolidation treatment of 107 patients with MK-AML consisted of alloHSCT (n = 45), chemotherapy (n = 48), or autologous HSCT (n = 14). RESULTS The 5-year overall survival after start of consolidation was 19% for patients with MK-AML who received alloHSCT and 9% for those who received chemotherapy or autoHSCT (P = .02). Relapse-free survival (RFS) at 5 years was 17% versus 7% (P = .003). Cox regression analysis was performed with alloHSCT as a time-dependent covariate. Hazard ratios (HRs) associated with alloHSCT for relapse and RFS were 0.30 (95% CI, 0.24 to 0.37; P < .001), and 0.52 (95% CI, 0.43 to 0.62; P < .001), respectively. HRs were similar in MK-AML and the other cytogenetic subgroups. CONCLUSION AlloHSCT, applied as consolidation in CR1, is associated with a significant reduction of relapse and improvement of survival in MK-AML, with the same relative reduction of relapse or death as in other cytogenetic risk categories.
Collapse
|
87
|
Kim Y, Yoon S, Kim SJ, Kim JS, Cheong JW, Min YH. Myeloperoxidase expression in acute myeloid leukemia helps identifying patients to benefit from transplant. Yonsei Med J 2012; 53:530-6. [PMID: 22476996 PMCID: PMC3343448 DOI: 10.3349/ymj.2012.53.3.530] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
PURPOSE Despite extensive study, the use of allogeneic hematopoietic stem cell transplantation in acute myeloid leukemia (AML) vary considerably. The decision of which of these options to choose is complex and depends on both clinical and molecular variables as well as the availability and histocompatability of donor stem cells. So far there is no clear explanation on whether the expression of myeloperoxidase (MPO) relates to the prognosis of AML. MATERIALS AND METHODS We retrospectively analyzed the prognostic significance of the MPO expression in the 140 patients with diagnosed AML treated at a single institution. RESULTS In our study, MPO expression was associated with disease-free survival (DFS) and transplant was beneficial to overcome a negative prognostic effect of MPO-negative at diagnosis based upon the result that the DFS in patients received transplants are not significant between the MPO-positive group and MPO-negative group although DFS in all patients was different according to MPO expression. CONCLUSION MPO expression at diagnosis helps to choose therapy for each AML patient and can differentiate AML patients who need transplantation.
Collapse
Affiliation(s)
- Yundeok Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Sulhee Yoon
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Soo Jeong Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jin Seok Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jun-Won Cheong
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Yoo Hong Min
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
88
|
Abstract
The prognostic factors in acute leukemia have undergone a major change over the past decade and are likely to be further refined in the coming years. While age is the single most important prognostic factor in both AML and in ALL, recurring cytogenetic abnormalities and molecular markers have become crucial for the prognosis of patients and for new directions in the development of targeted therapies. No less important is the development of a personalized approach for therapy as determined by the response to therapy using increasingly sensitive technologies. The assessment of MRD is rapidly superseding other prognostic factors in ALL and, somewhat lacking behind, coming into its own in AML. The next decade should see further refinement of response-driven prognostication, to include epigenetics as well as pharmacogenetics and pharmacodynamics of individual drugs used and the responses to them. It is hoped that these refinements and better predictors of response will also lead to a significantly improved overall outcome of patients with both AML and ALL.
Collapse
Affiliation(s)
- Chezi Ganzel
- Department of Hematology, Shaare Zedek Medical Center, PO Box 3235, Jerusalem 91031, Israel.
| | | |
Collapse
|
89
|
Grimwade D, Mrózek K. Diagnostic and prognostic value of cytogenetics in acute myeloid leukemia. Hematol Oncol Clin North Am 2012; 25:1135-61, vii. [PMID: 22093581 DOI: 10.1016/j.hoc.2011.09.018] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The last 4 decades have seen major advances in understanding the genetic basis of acute myeloid leukemia (AML), and substantial improvements in survival of children and young adults with the disease. A key step forward was the discovery that AML cells harbor recurring cytogenetic abnormalities. The identification of the genes involved in chromosomal rearrangements has provided insights into the regulation of normal hematopoiesis and how disruption of key transcription factors and epigenetic modulators promote leukemic transformation. Cytogenetics has been widely adopted to provide the framework for development of risk-stratified treatment approaches to patient management.
Collapse
Affiliation(s)
- David Grimwade
- Cancer Genetics Laboratory, Department of Medical & Molecular Genetics, Guy's Hospital, King's College London School of Medicine, 8th Floor, Guy's Tower, London SE1 9RT, UK.
| | | |
Collapse
|
90
|
Miglino M, Colombo N, Pica G, Grasso R, Clavio M, Bergamaschi M, Ballerini F, Ghiso A, Ghiggi C, Mitscheunig L, Beltrami G, Cagnetta A, Vignolo L, Lucchetti MV, Aquino S, Pierri I, Sessarego M, Carella AM, Gobbi M. WT1 overexpression at diagnosis may predict favorable outcome in patients with de novo non-M3 acute myeloid leukemia. Leuk Lymphoma 2012; 52:1961-9. [PMID: 21942328 DOI: 10.3109/10428194.2011.585673] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We reviewed the frequency and prognostic significance of FLT3 (fms-like tyrosine kinase receptor-3) and NPM (nucleophosmin) gene mutations and WT1 (Wilms' tumor) and BAALC (brain and acute leukemia, cytoplasmic) gene expression in 100 consecutive patients with intermediate and poor cytogenetic risk de novo acute myeloid leukemia (AML) receiving conventional anthracycline-AraC based therapy. We observed a strict relationship between unfavorable karyotype and BAALC >1000 (p = 0.0001). Multivariate analysis of 81 patients with intermediate karyotype revealed that younger age (p = 0.00009), NPM gene mutation (p = 0.002), and WT1 >75th percentile (>2365) (p = 0.003) were independent, positive factors for complete remission (CR). WT1 expression above 2365 was correlated also to longer event-free survival (EFS) and overall survival (OS) in the same subset of patients (p = 0.003 and p = 0.02, respectively); the same finding occurred in younger patients with AML with intermediate karyotype (p = 0.008 and p = 0.01, respectively). In patients with intermediate karyotype, FLT3 internal tandem duplication (ITD) negatively affected EFS (EFS at 30 months: 30% vs. 6% in FLT3-ITD negative and FLT3 positive patients, respectively; p = 0.01) and OS (OS at 30 months: 38% vs. 20%, p = 0.03). The positive prognostic value of high WT1 expression does not have a clear explanation; it may be implicated either with WT1 anti-oncogenic function, or with the stimulating effect of WT1 oncogene on the leukemic cellular cycle, possibly associated with an enhanced response to chemotherapy.
Collapse
Affiliation(s)
- Maurizio Miglino
- Department of Hematology and Oncology, Azienda Ospedaliera Universitaria San Martino, Genova, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Bullinger L, Fröhling S. Array-Based Cytogenetic Approaches in Acute Myeloid Leukemia: Clinical Impact and Biological Insights. Semin Oncol 2012; 39:37-46. [DOI: 10.1053/j.seminoncol.2011.11.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
92
|
Azacitidine augments expansion of regulatory T cells after allogeneic stem cell transplantation in patients with acute myeloid leukemia (AML). Blood 2012; 119:3361-9. [PMID: 22234690 DOI: 10.1182/blood-2011-09-377044] [Citation(s) in RCA: 321] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Strategies that augment a GVL effect without increasing the risk of GVHD are required to improve the outcome after allogeneic stem cell transplantation (SCT). Azacitidine (AZA) up-regulates the expression of tumor Ags on leukemic blasts in vitro and expands the numbers of immunomodulatory T regulatory cells (Tregs) in animal models. Reasoning that AZA might selectively augment a GVL effect, we studied the immunologic sequelae of AZA administration after allogeneic SCT. Twenty-seven patients who had undergone a reduced intensity allogeneic transplantation for acute myeloid leukemia were treated with monthly courses of AZA, and CD8(+) T-cell responses to candidate tumor Ags and circulating Tregs were measured. AZA after transplantation was well tolerated, and its administration was associated with a low incidence of GVHD. Administration of AZA increased the number of Tregs within the first 3 months after transplantation compared with a control population (P = .0127). AZA administration also induced a cytotoxic CD8(+) T-cell response to several tumor Ags, including melanoma-associated Ag 1, B melanoma antigen 1, and Wilm tumor Ag 1. These data support the further examination of AZA after transplantation as a mechanism of augmenting a GVL effect without a concomitant increase in GVHD.
Collapse
|
93
|
Singh R, Cadeddu RP, Fröbel J, Wilk CM, Bruns I, Zerbini LF, Prenzel T, Hartwig S, Brünnert D, Schroeder T, Lehr S, Haas R, Czibere A. The non-steroidal anti-inflammatory drugs Sulindac sulfide and Diclofenac induce apoptosis and differentiation in human acute myeloid leukemia cells through an AP-1 dependent pathway. Apoptosis 2011; 16:889-901. [PMID: 21739277 DOI: 10.1007/s10495-011-0624-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Acute myeloid leukemia is a heterogeneous disease with varying genetic and molecular pathologies. Non-steroidal anti-inflammatory drugs (NSAIDs) have been proven to possess significant anti-proliferative potential in various cancer cells in vitro and in vivo. Hence, treatment with these agents can be utilized to study disease specific anti-proliferative pathways. In this study, a total number of 42 bone marrow derived CD34(+) selected de novo AML patient samples and the AML cell lines THP-1 and HL-60 were treated with the NSAIDs Sulindac sulfide and Diclofenac. We analyzed viability, apoptosis, differentiation and addressed the molecular mechanisms involved. We found a consistent induction of apoptosis and to some extent an increased myeloid differentiation capacity in NSAID treated AML cells. Comprehensive protein and gene expression profiling of Diclofenac treated AML cells revealed transcriptional activation of GADD45α and its downstream MAPK/JNK pathway as well as increased protein levels of the caspase-3 precursor. This pointed towards a role of the c-Jun NH(2)-terminal kinase (JNK) in NSAID mediated apoptosis that we found indeed to be dependent on JNK activity as addition of a specific JNK-inhibitor abrogated apoptosis. Furthermore, the AP-1 transcription factor family members' c-Jun, JunB and Fra-2 were transcriptionally activated in NSAID treated AML cells and re-expression of these transcription factors led to activation of GADD45α with induction of apoptosis. Mechanistically, we demonstrate that NSAIDs induce apoptosis in AML through a novel pathway involving increased expression of AP-1 heterodimers, which by itself is sufficient to induce GADD45α expression with consecutive activation of JNK and induction of apoptosis.
Collapse
Affiliation(s)
- Raminder Singh
- Department of Hematology, Oncology and Clinical Immunology, Heinrich Heine-University, Düsseldorf, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Amplification of c-MYC and MLL Genes as a Marker of Clonal Cell Progression in Patients with Myeloid Malignancy and Trisomy of Chromosomes 8 or 11. Balkan J Med Genet 2011; 14:17-24. [PMID: 24052708 PMCID: PMC3776705 DOI: 10.2478/v10034-011-0043-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Gene amplification (amp) is one of the basic mechanisms connected with overexpression of oncogenes. The c-MYC (located in 8q24) and MLL (located in 11q23) are the most often over represented genes that lead to a rapid proliferation of the affected cell clone in patients with myeloid neoplasms. Assessment of the level of amp c-MYC or amp MLL in the cases with trisomy 8 (+8) or trisomy 11 (+11) and myeloid malignances is necessary for a more precise estimation of the disease progression. A total of 26 patients with acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) were included in the study: 18 with +8, six with +11 and two with complex karyotypes suspected of the partial trisomy. Routine cytogenetic analysis and fluorescent in situ hybridization (FISH) were applied to indicate the chromosome alterations and genes amp in the bone marrow cells. Amp c-MYC was observed in 12 from 18 (66.7%) patients with +8. All the patients with +11 demonstrated a different level of amp MLL. In most of the cases with MDS (9/10), the coincidence of the +8 or +11 with amp c-MYC or amp MLL, respectively, leads to transformation to AML and/or short overall survival. Our data suggest that amp c-MYC and amp MLL develop in conformity with +8 and +11, especially in cases with progressive deviations in the karyotype as an aggressive expansion of an aberrant cell clone and appearance of additional chromosome anomalies.
Collapse
|
95
|
Löwenberg B, Muus P, Ossenkoppele G, Rousselot P, Cahn JY, Ifrah N, Martinelli G, Amadori S, Berman E, Sonneveld P, Jongen-Lavrencic M, Rigaudeau S, Stockman P, Goudie A, Faderl S, Jabbour E, Kantarjian H. Phase 1/2 study to assess the safety, efficacy, and pharmacokinetics of barasertib (AZD1152) in patients with advanced acute myeloid leukemia. Blood 2011; 118:6030-6. [PMID: 21976672 PMCID: PMC4186639 DOI: 10.1182/blood-2011-07-366930] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 09/27/2011] [Indexed: 11/20/2022] Open
Abstract
The primary objective of this 2-part phase 1/2 study was to determine the maximum-tolerated dose (MTD) of the potent and selective Aurora B kinase inhibitor barasertib (AZD1152) in patients with newly diagnosed or relapsed acute myeloid leukemia (AML). Part A determined the MTD of barasertib administered as a continuous 7-day infusion every 21 days. In part B, the efficacy of barasertib was evaluated at the MTD. In part A, 32 patients were treated with barasertib 50 mg (n = 3), 100 mg (n = 3), 200 mg (n = 3), 400 mg (n = 4), 800 mg (n = 7), 1200 mg (n = 6), and 1600 mg (n = 6). Dose-limiting toxicities (stomatitis/mucosal inflammation events) were reported in the 800 mg (n = 1), 1200 mg (n = 1), and 1600 mg (n = 2) groups. The MTD was defined as 1200 mg. In part B, 32 patients received barasertib 1200 mg. In each part of the study, 8 of 32 patients had a hematologic response according to Cheson AML criteria. The most commonly reported grade ≥ 3 events were febrile neutropenia (n = 24) and stomatitis/mucosal inflammation (n = 16). We concluded that the MTD of barasertib is 1200 mg in patients with relapsed or newly diagnosed AML. Toxicity was manageable and barasertib treatment resulted in an overall hematologic response rate of 25%. This study is registered at www.ClinicalTrials.gov as NCT00497991.
Collapse
Affiliation(s)
- Bob Löwenberg
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Somatic mutations in the transcriptional corepressor gene BCORL1 in adult acute myelogenous leukemia. Blood 2011; 118:5914-7. [PMID: 21989985 DOI: 10.1182/blood-2011-05-356204] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
To further our understanding of the genetic basis of acute myelogenous leukemia (AML), we determined the coding exon sequences of ∼ 18 000 protein-encoding genes in 8 patients with secondary AML. Here we report the discovery of novel somatic mutations in the transcriptional corepressor gene BCORL1 that is located on the X-chromosome. Analysis of BCORL1 in an unselected cohort of 173 AML patients identified a total of 10 mutated cases (6%) with BCORL1 mutations, whereas analysis of 19 AML cell lines uncovered 4 (21%) BCORL1 mutated cell lines. The majority (87%) of the mutations in BCORL1 were predicted to inactivate the gene product as a result of nonsense mutations, splice site mutation, or out-of-frame insertions or deletions. These results indicate that BCORL1 by genetic criteria is a novel candidate tumor suppressor gene, joining the growing list of genes recurrently mutated in AML.
Collapse
|
97
|
Brown T, Swansbury J, Taj MM. Prognosis of patients with t(8;16)(p11;p13) acute myeloid leukemia. Leuk Lymphoma 2011; 53:338-41. [PMID: 21846182 DOI: 10.3109/10428194.2011.614703] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
MESH Headings
- Adolescent
- Adult
- Child
- Child, Preschool
- Chromosomes, Human, Pair 16/genetics
- Chromosomes, Human, Pair 8/genetics
- Female
- Humans
- Infant
- Infant, Newborn
- Karyotyping
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/therapy
- Male
- Prognosis
- Survival Rate
- Translocation, Genetic/genetics
- Young Adult
Collapse
|
98
|
Larsen HØ, Roug AS, Just T, Brown GD, Hokland P. Expression of the hMICL in acute myeloid leukemia-a highly reliable disease marker at diagnosis and during follow-up. CYTOMETRY PART B-CLINICAL CYTOMETRY 2011; 82:3-8. [PMID: 22173921 DOI: 10.1002/cyto.b.20614] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 07/24/2011] [Accepted: 07/25/2011] [Indexed: 12/21/2022]
Abstract
BACKGROUND Stable flow cytometric markers for malignant myeloid cells are highly warranted. Based on data from stem cell research, we hypothesized that the human inhibitory C-type lectin like receptor (hMICL) might represent a novel diagnostic and prognostic vehicle in a standard flow cytometry (FCM) setting. METHODS Standard four-color FCM was employed to uncover the expression patterns of hMICL in bone marrow in a test set of 55 retrospectively collected diagnostic acute myeloid leukemia (AML) samples and in a set of 36 prospectively collected diagnostic AML samples. RESULTS Ninety-two percent of the AML patients stained positive for hMICL and in the otherwise poorly characterized CD34 negative patient group hMICL staining revealed a very homogenous expression profile in the blast cell compartment with a mean of 88% hMICL positive cells. Moreover, hMICL displayed significantly higher expression in AML as compared with normal donors as measured by median fluorescence intensity (MFI) ratios (P = 0.01). There was no difference in hMICL MFI ratios between the CD34 positive and the CD34 negative subgroups (P = 0.89). Importantly, there was no difference in MFI ratios between paired diagnostic and relapse samples (P = 0.76) in 23 cases studied, indicating stable expression of hMICL during the course of the disease. In contrast to the other stem cell associated antigens analyzed (CD34, CD96, CD117, and CD133), hMICL was expressed on myeloid blast cells only, revealing hMICL as a diagnostic marker in AML. CONCLUSION These data identify hMICL as a myeloid leukemia-associated antigen and establishes its applicability for diagnosis and follow-up of AML patients in a standard FCM setting.
Collapse
Affiliation(s)
- Hanne Ø Larsen
- Department of Hematology, The Laboratory of Immunohematology, Aarhus University Hospital, Aarhus, Denmark
| | | | | | | | | |
Collapse
|
99
|
Maurillo L, Venditti A, Spagnoli A, Gaidano G, Ferrero D, Oliva E, Lunghi M, D'Arco AM, Levis A, Pastore D, Di Renzo N, Santagostino A, Pavone V, Buccisano F, Musto P. Azacitidine for the treatment of patients with acute myeloid leukemia. Cancer 2011; 118:1014-22. [DOI: 10.1002/cncr.26354] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Revised: 04/27/2011] [Accepted: 05/20/2011] [Indexed: 11/10/2022]
|
100
|
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: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|