Your browser doesn't support javascript.
loading
Immune Reconstitution following High-Dose Chemotherapy and Autologous Stem Cell Transplantation with or without Pembrolizumab Maintenance Therapy in Patients with Lymphoma.
Merryman, Reid W; Redd, Robert; Jeter, Erin; Wong, Jeff L; McHugh, Kristin; Reynolds, Carol; Nazzaro, Matthew; Varden, Aine; Brown, Jennifer R; Crombie, Jennifer L; Davids, Matthew S; Fisher, David C; Jacobsen, Eric; Jacobson, Caron A; Kim, Austin I; LaCasce, Ann S; Ng, Samuel Y; Odejide, Oreofe O; Parry, Erin M; Dahi, Parastoo B; Nieto, Yago; Joyce, Robin M; Chen, Yi-Bin; Herrera, Alex F; Armand, Philippe; Ritz, Jerome.
Afiliação
  • Merryman RW; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Electronic address: Reid_merryman@dfci.harvard.edu.
  • Redd R; Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Jeter E; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Wong JL; Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, New York; Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan-Kettering Cancer Center, New York, New York.
  • McHugh K; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Reynolds C; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Nazzaro M; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Varden A; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Brown JR; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Crombie JL; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Davids MS; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Fisher DC; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Jacobsen E; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Jacobson CA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Kim AI; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • LaCasce AS; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Ng SY; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Odejide OO; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Parry EM; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Dahi PB; Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan-Kettering Cancer Center, New York, New York.
  • Nieto Y; Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Joyce RM; Department of Hematologic Malignancy, Beth Israel Deaconess Medical Center, Boston, Massachusetts.
  • Chen YB; Bone Marrow Transplantation Program, Massachusetts General Hospital, Boston, Massachusetts.
  • Herrera AF; Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California.
  • Armand P; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Ritz J; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
Transplant Cell Ther ; 28(1): 32.e1-32.e10, 2022 01.
Article em En | MEDLINE | ID: mdl-34670169
ABSTRACT
Autologous stem cell transplantation (ASCT) is a standard of care for patients with chemosensitive, relapsed/refractory (R/R) classical Hodgkin lymphoma (cHL) and diffuse large B cell lymphoma (DLBCL). Whereas the clinical benefit of ASCT has traditionally been attributed solely to cytoreduction from intensive chemotherapy, ASCT has important immunogenic effects that may contribute to its antitumor efficacy and could provide a favorable immune environment for post-ASCT immune-based maintenance treatments. We previously reported clinical results of a phase II trial (ClinicalTrials.gov identifier NCT02362997) testing 8 doses of pembrolizumab maintenance therapy after ASCT for patients with R/R cHL or DLBCL. To clarify the impact of pembrolizumab on immune reconstitution, we compared the kinetics of peripheral blood immune cell recovery after ASCT for trial patients receiving pembrolizumab maintenance to those of a contemporaneous control cohort of similar patients undergoing ASCT without pembrolizumab maintenance. This study was conducted to characterize the impact of post-ASCT pembrolizumab maintenance therapy on immune reconstitution for patients with R/R DLBCL and cHL and to identify candidate biomarkers of efficacy and immune-related adverse events (irAEs). Peripheral blood (PB) mononuclear cell samples were prospectively collected at 1 to 18 months after ASCT and analyzed by flow cytometry using a panel of fluorophore-conjugated monoclonal antibodies to identify B cells, natural killer (NK) cells, and various dendritic cell (DC) and T cell subsets. A median of 5 (range, 1 to 8) post-ASCT PB samples were collected from 144 patients (59 in the pembrolizumab group and 85 in the control group). Clinical characteristics of the 2 cohorts were similar. Compared with cHL patients, DLBCL patients (all of whom received anti-CD20 monoclonal antibody therapy before ASCT) had delayed CD19+ cell reconstitution that persisted for at least 18 months after ASCT. No other differences in immune reconstitution based on lymphoma subtype were observed. Post-ASCT pembrolizumab maintenance therapy was associated with an elevation in circulating DCs (driven by higher levels of plasmacytoid and immature DCs) that persisted for the duration of pembrolizumab treatment, along with a significant reduction in PD-1+ T cells that persisted for 6 to 12 months after completion of pembrolizumab therapy. Despite the key role of T cells in mediating the effects of PD-1 blockade, pembrolizumab maintenance did not affect recovery of any T cell subsets. In an exploratory analysis, a higher baseline CD4+ terminal effector memory cell count (defined as CD3+CD4+CD45RA+CD62L-) was associated with inferior progression-free survival (PFS), but only among patients who received pembrolizumab maintenance (P = .003). As continuous variables, lower absolute levels of NK cells (P = .009), PD-1+ CD4+ T cells (P = .005), and PD-1+ CD8+ T cells (P = .005) before pembrolizumab initiation were each associated with a higher risk of grade 2+ irAEs. Our findings indicate that post-ACST pembrolizumab maintenance therapy is associated with a persistent elevation of circulating DCs, but its impact on the reconstitution of other immune cells in peripheral blood appears limited. Our study suggests that early features of post-ASCT immune reconstitution could be associated with PFS and the risk of irAE and warrant additional investigation. © 2021 American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Doença de Hodgkin / Transplante de Células-Tronco Hematopoéticas / Reconstituição Imune Tipo de estudo: Prognostic_studies Limite: Humans Idioma: En Revista: Transplant Cell Ther Ano de publicação: 2022 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Doença de Hodgkin / Transplante de Células-Tronco Hematopoéticas / Reconstituição Imune Tipo de estudo: Prognostic_studies Limite: Humans Idioma: En Revista: Transplant Cell Ther Ano de publicação: 2022 Tipo de documento: Article