Your browser doesn't support javascript.
loading
Impact of diagnostic genetics on remission MRD and transplantation outcomes in older patients with AML.
Murdock, H Moses; Kim, Haesook T; Denlinger, Nathan; Vachhani, Pankit; Hambley, Bryan; Manning, Bryan S; Gier, Shannon; Cho, Christina; Tsai, Harrison K; McCurdy, Shannon; Ho, Vincent T; Koreth, John; Soiffer, Robert J; Ritz, Jerome; Carroll, Martin P; Vasu, Sumithira; Perales, Miguel-Angel; Wang, Eunice S; Gondek, Lukasz P; Devine, Steven; Alyea, Edwin P; Lindsley, R Coleman; Gibson, Christopher J.
Afiliación
  • Murdock HM; Division of Hematologic Neoplasia, Department of Medical Oncology, and.
  • Kim HT; Department of Data Science, Dana-Farber Cancer Institute, Boston, MA.
  • Denlinger N; Division of Hematology, The Ohio State University James Cancer Hospital, Columbus, OH.
  • Vachhani P; Division of Hematology and Oncology, University of Alabama at Birmingham School of Medicine, Birmingham, AL.
  • Hambley B; Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH.
  • Manning BS; Department of Medicine, Perelman Cancer Center, University of Pennsylvania, Philadelphia, PA.
  • Gier S; Department of Medicine, Perelman Cancer Center, University of Pennsylvania, Philadelphia, PA.
  • Cho C; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY.
  • Tsai HK; Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA.
  • McCurdy S; Department of Medicine, Perelman Cancer Center, University of Pennsylvania, Philadelphia, PA.
  • Ho VT; Division of Hematologic Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA.
  • Koreth J; Division of Hematologic Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA.
  • Soiffer RJ; Division of Hematologic Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA.
  • Ritz J; Division of Hematologic Neoplasia, Department of Medical Oncology, and.
  • Carroll MP; Department of Medicine, Perelman Cancer Center, University of Pennsylvania, Philadelphia, PA.
  • Vasu S; Division of Hematology, The Ohio State University James Cancer Hospital, Columbus, OH.
  • Perales MA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY.
  • Wang ES; Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY.
  • Gondek LP; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD.
  • Devine S; National Marrow Donor Program, Minneapolis, MN; and.
  • Alyea EP; Duke Cancer Institute, Duke University Medical Center, Durham, NC.
  • Lindsley RC; Division of Hematologic Neoplasia, Department of Medical Oncology, and.
  • Gibson CJ; Division of Hematologic Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA.
Blood ; 139(24): 3546-3557, 2022 06 16.
Article en En | MEDLINE | ID: mdl-35286378
ABSTRACT
Older patients with acute myeloid leukemia (AML) have high relapse risk and poor survival after allogeneic hematopoietic cell transplantation (HCT). Younger patients may receive myeloablative conditioning to mitigate relapse risk associated with high-risk genetics or measurable residual disease (MRD), but older adults typically receive reduced-intensity conditioning (RIC) to limit toxicity. To identify factors that drive HCT outcomes in older patients, we performed targeted mutational analysis (variant allele fraction ≥2%) on diagnostic samples from 295 patients with AML aged ≥60 years who underwent HCT in first complete remission, 91% of whom received RIC, and targeted duplex sequencing at remission in a subset comprising 192 patients. In a multivariable model for leukemia-free survival (LFS) including baseline genetic and clinical variables, we defined patients with low (3-year LFS, 85%), intermediate (55%), high (35%), and very high (7%) risk. Before HCT, 79.7% of patients had persistent baseline mutations, including 18.3% with only DNMT3A or TET2 (DT) mutations and 61.4% with other mutations (MRD positive). In univariable analysis, MRD positivity was associated with increased relapse and inferior LFS, compared with DT and MRD-negative mutations. However, in a multivariable model accounting for baseline risk, MRD positivity had no independent impact on LFS, most likely because of its significant association with diagnostic genetic characteristics, including MDS-associated gene mutations, TP53 mutations, and high-risk karyotype. In summary, molecular associations with MRD positivity and transplant outcomes in older patients with AML are driven primarily by baseline genetics, not by mutations present in remission. In this group of patients, where high-intensity conditioning carries substantial risk of toxicity, alternative approaches to mitigating MRD-associated relapse risk are needed.
Asunto(s)

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Leucemia Mieloide Aguda / Trasplante de Células Madre Hematopoyéticas Tipo de estudio: Diagnostic_studies / Observational_studies / Prognostic_studies Límite: Aged / Humans Idioma: En Año: 2022 Tipo del documento: Article

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Leucemia Mieloide Aguda / Trasplante de Células Madre Hematopoyéticas Tipo de estudio: Diagnostic_studies / Observational_studies / Prognostic_studies Límite: Aged / Humans Idioma: En Año: 2022 Tipo del documento: Article