1
|
Braidotti S, Granzotto M, Curci D, Faganel Kotnik B, Maximova N. Advancing Allogeneic Hematopoietic Stem Cell Transplantation Outcomes through Immunotherapy: A Comprehensive Review of Optimizing Non-CAR Donor T-Lymphocyte Infusion Strategies. Biomedicines 2024; 12:1853. [PMID: 39200317 PMCID: PMC11351482 DOI: 10.3390/biomedicines12081853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/07/2024] [Accepted: 08/11/2024] [Indexed: 09/02/2024] Open
Abstract
Optimized use of prophylactic or therapeutic donor lymphocyte infusions (DLI) is aimed at improving clinical outcomes in patients with malignant and non-malignant hematological diseases who have undergone allogeneic hematopoietic stem cell transplantation (allo-HSCT). Memory T-lymphocytes (CD45RA-/CD45RO+) play a crucial role in immune reconstitution post-HSCT. The infusion of memory T cells is proven to be safe and effective in improving outcomes due to the enhanced reconstitution of immunity and increased protection against viremia, without exacerbating graft-versus-host disease (GVHD) risks. Studies indicate their persistence and efficacy in combating viral pathogens, suggesting a viable therapeutic avenue for patients. Conversely, using virus-specific T cells for viremia control presents challenges, such as regulatory hurdles, cost, and production time compared to CD45RA-memory T lymphocytes. Additionally, the modulation of regulatory T cells (Tregs) for therapeutic use has become an important area of investigation in GVHD, playing a pivotal role in immune tolerance modulation, potentially mitigating GVHD and reducing pharmacological immunosuppression requirements. Finally, donor T cell-mediated graft-versus-leukemia immune responses hold promise in curbing relapse rates post-HSCT, providing a multifaceted approach to therapeutic intervention in high-risk disease scenarios. This comprehensive review underscores the multifaceted roles of T lymphocytes in HSCT outcomes and identifies avenues for further research and clinical application.
Collapse
Affiliation(s)
- Stefania Braidotti
- Department of Pediatrics, Institute for Maternal and Child Health-IRCCS Burlo Garofolo, 34137 Trieste, Italy;
| | - Marilena Granzotto
- Azienda Sanitaria Universitaria Giuliano Isontina (ASU GI), 34125 Trieste, Italy;
| | - Debora Curci
- Advanced Translational Diagnostic Laboratory, Institute for Maternal and Child Health-IRCCS Burlo Garofolo, 34137 Trieste, Italy;
| | - Barbara Faganel Kotnik
- Department of Hematology and Oncology, University Children’s Hospital, 1000 Ljubljana, Slovenia;
| | - Natalia Maximova
- Department of Pediatrics, Institute for Maternal and Child Health-IRCCS Burlo Garofolo, 34137 Trieste, Italy;
| |
Collapse
|
2
|
Lamba DS, Tripathi PP, Hans R, Khadwal A, Sharma RR. Donor lymphocyte infusions: An experience from a tertiary care center of North India. Asian J Transfus Sci 2024; 18:124-127. [PMID: 39036678 PMCID: PMC11259343 DOI: 10.4103/ajts.ajts_211_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/11/2024] [Accepted: 03/03/2024] [Indexed: 07/23/2024] Open
Abstract
Donor lymphocyte infusions (DLIs) are often recommended products after allogeneic hematopoietic stem cell transplant to increase graft - versus - leukemia effect. More success rate of DLI has been reported in relapsed posttransplant chronic myeloid leukemia. Whatever the indication for DLI, mortality related to post-DLI infusion is 5%-20%, and more than one-third of patients will develop acute and/or chronic graft versus host disease (GVHD) after DLI. We report two cases where DLIs were used for residual disease after posttransplant. Both of DLI went uneventful. None of the patient's developed signs of GVHD postinfusion. Although both patients expired with different causes, none were related to DLI infusion. Information from published literature suggests that DLI should be administered early after relapse or as a prophylactic strategy in patients receiving T-cell-depleted grafts, and patients with aggressive diseases may benefit from disease reduction before DLI. However, further evidence is required to evaluate its efficacy, especially in relapsed or residual hematological malignancies.
Collapse
Affiliation(s)
- Divjot Singh Lamba
- Department of Transfusion Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Parmatma Prasad Tripathi
- Department of Transfusion Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Rekha Hans
- Department of Transfusion Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Alka Khadwal
- Department of Clinical Hematology and Medical Oncology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Ratti Ram Sharma
- Department of Transfusion Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
3
|
Riva C, Vernarecci C, Minetto P, Goda R, Greppi M, Pesce S, Chies M, Zecchetti G, Ferro B, Maio E, Cea M, Lemoli RM, Marcenaro E, Guolo F. Harnessing Immune Response in Acute Myeloid Leukemia. J Clin Med 2023; 12:5824. [PMID: 37762763 PMCID: PMC10532363 DOI: 10.3390/jcm12185824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 08/30/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Despite the results achieved with the evolution of conventional chemotherapy and the inclusion of targeted therapies in the treatment of acute myeloid leukemia (AML), survival is still not satisfying, in particular in the setting of relapsed/refractory (R/R) disease or elderly/unfit patients. Among the most innovative therapeutic options, cellular therapy has shown great results in different hematological malignancies such as acute lymphoblastic leukemia and lymphomas, with several products already approved for clinical use. However, despite the great interest in also expanding the application of these new treatments to R/R AML, no product has been approved yet for clinical application. Furthermore, cellular therapy could indeed represent a powerful tool and an appealing alternative to allogeneic hematopoietic stem cell transplantation for ineligible patients. In this review, we aim to provide an overview of the most recent clinical research exploring the effectiveness of cellular therapy in AML, moving from consolidated approaches such as post- transplant donor's lymphocytes infusion, to modern adoptive immunotherapies such as alloreactive NK cell infusions, engineered T and NK cells (CAR-T, CAR-NK) and novel platforms of T and NK cells engaging (i.e., BiTEs, DARTs and ANKETTM).
Collapse
Affiliation(s)
- Carola Riva
- Clinic of Hematology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy; (C.R.); (C.V.); (M.C.); (G.Z.); (B.F.); (E.M.); (M.C.); (R.M.L.); (F.G.)
| | - Chiara Vernarecci
- Clinic of Hematology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy; (C.R.); (C.V.); (M.C.); (G.Z.); (B.F.); (E.M.); (M.C.); (R.M.L.); (F.G.)
| | - Paola Minetto
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Rayan Goda
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (R.G.); (M.G.); (S.P.)
| | - Marco Greppi
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (R.G.); (M.G.); (S.P.)
| | - Silvia Pesce
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (R.G.); (M.G.); (S.P.)
| | - Maria Chies
- Clinic of Hematology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy; (C.R.); (C.V.); (M.C.); (G.Z.); (B.F.); (E.M.); (M.C.); (R.M.L.); (F.G.)
| | - Giada Zecchetti
- Clinic of Hematology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy; (C.R.); (C.V.); (M.C.); (G.Z.); (B.F.); (E.M.); (M.C.); (R.M.L.); (F.G.)
| | - Beatrice Ferro
- Clinic of Hematology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy; (C.R.); (C.V.); (M.C.); (G.Z.); (B.F.); (E.M.); (M.C.); (R.M.L.); (F.G.)
| | - Elena Maio
- Clinic of Hematology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy; (C.R.); (C.V.); (M.C.); (G.Z.); (B.F.); (E.M.); (M.C.); (R.M.L.); (F.G.)
| | - Michele Cea
- Clinic of Hematology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy; (C.R.); (C.V.); (M.C.); (G.Z.); (B.F.); (E.M.); (M.C.); (R.M.L.); (F.G.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Roberto Massimo Lemoli
- Clinic of Hematology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy; (C.R.); (C.V.); (M.C.); (G.Z.); (B.F.); (E.M.); (M.C.); (R.M.L.); (F.G.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Emanuela Marcenaro
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (R.G.); (M.G.); (S.P.)
| | - Fabio Guolo
- Clinic of Hematology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy; (C.R.); (C.V.); (M.C.); (G.Z.); (B.F.); (E.M.); (M.C.); (R.M.L.); (F.G.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| |
Collapse
|
4
|
Hodroj MH, Abou Dalle I, Moukalled N, El Cheikh J, Mohty M, Bazarbachi A. Novel strategies to prevent and overcome relapse after allogeneic hematopoietic cell transplantation in acute lymphoblastic leukemia. Front Immunol 2023; 14:1191912. [PMID: 37359547 PMCID: PMC10285443 DOI: 10.3389/fimmu.2023.1191912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/26/2023] [Indexed: 06/28/2023] Open
Abstract
The outcome of B-cell acute lymphoblastic leukemia (B-ALL) has improved over time with the incorporation of multi-agent chemotherapy in the treatment landscape as well as the recent approval of immunotherapeutic agents allowing a larger proportion of patients to undergo allogeneic hematopoietic cell transplantation (allo-HCT) which is still considered a potential curative approach. However, relapse post-transplant is still occurring and constitutes a common cause of treatment failure in B-ALL. The present review aims to discuss the novel strategies and therapies used to prevent and overcome relapse post allo-HCT in patients with ALL, focusing on the role of tyrosine kinase inhibitors in Philadelphia chromosome positive B-ALL, the role of innovative agents such as blinatumomab and inotuzumab ozogamicin, and finally the role of cellular therapy.
Collapse
Affiliation(s)
- Mohammad Hassan Hodroj
- Division of Hematology & Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Iman Abou Dalle
- Division of Hematology & Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
- Bone Marrow Transplantation Program, American University of Beirut Medical Center, Beirut, Lebanon
| | - Nour Moukalled
- Division of Hematology & Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
- Bone Marrow Transplantation Program, American University of Beirut Medical Center, Beirut, Lebanon
| | - Jean El Cheikh
- Division of Hematology & Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
- Bone Marrow Transplantation Program, American University of Beirut Medical Center, Beirut, Lebanon
| | - Mohamad Mohty
- Sorbonne University, Saint-Antoine Hospital, AP-HP, INSERM UMRs 938, Paris, France
| | - Ali Bazarbachi
- Division of Hematology & Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
- Bone Marrow Transplantation Program, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
5
|
Harada K. Pre-emptive and prophylactic donor lymphocyte infusion following allogeneic stem cell transplantation. Int J Hematol 2023:10.1007/s12185-023-03595-x. [PMID: 37014602 DOI: 10.1007/s12185-023-03595-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 04/05/2023]
Abstract
Donor lymphocyte infusion (DLI) is an allogenic immunotherapy used after allogeneic hematopoietic stem cell transplantation. DLI takes advantage of the graft-versus-tumor effect induced by the infused CD3 + T cells, but may induce graft-versus-host disease. To date, DLI has been attempted to prevent hematological relapse after allogeneic hematopoietic stem cell transplantation in patients with mixed chimerism and molecular relapse (pre-emptive DLI), and as maintenance therapy in patients with high-risk hematological malignancies (prophylactic DLI). DLI response and efficacy depend on patient, disease, and DLI factors. This review discusses the efficacy and risks of DLI, with a focus on pre-emptive and prophylactic use.
Collapse
Affiliation(s)
- Kaito Harada
- Department of Hematology and Oncology, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1143, Japan.
| |
Collapse
|
6
|
Leotta S, Condorelli A, Sciortino R, Milone GA, Bellofiore C, Garibaldi B, Schininà G, Spadaro A, Cupri A, Milone G. Prevention and Treatment of Acute Myeloid Leukemia Relapse after Hematopoietic Stem Cell Transplantation: The State of the Art and Future Perspectives. J Clin Med 2022; 11:253. [PMID: 35011994 PMCID: PMC8745746 DOI: 10.3390/jcm11010253] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 12/24/2021] [Accepted: 12/28/2021] [Indexed: 12/19/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) for high-risk acute myeloid leukemia (AML) represents the only curative option. Progress has been made in the last two decades in the pre-transplant induction therapies, supportive care, selection of donors and conditioning regimens that allowed to extend the HSCT to a larger number of patients, including those aged over 65 years and/or lacking an HLA-identical donor. Furthermore, improvements in the prophylaxis of the graft-versus-host disease and of infection have dramatically reduced transplant-related mortality. The relapse of AML remains the major reason for transplant failure affecting almost 40-50% of the patients. From 10 to 15 years ago to date, treatment options for AML relapsing after HSCT were limited to conventional cytotoxic chemotherapy and donor leukocyte infusions (DLI). Nowadays, novel agents and targeted therapies have enriched the therapeutic landscape. Moreover, very recently, the therapeutic landscape has been enriched by manipulated cellular products (CAR-T, CAR-CIK, CAR-NK). In light of these new perspectives, careful monitoring of minimal-residual disease (MRD) and prompt application of pre-emptive strategies in the post-transplant setting have become imperative. Herein, we review the current state of the art on monitoring, prevention and treatment of relapse of AML after HSCT with particular attention on novel agents and future directions.
Collapse
Affiliation(s)
| | - Annalisa Condorelli
- Division of Hematology, AOU “Policlinico G. Rodolico-San Marco”, Via Santa Sofia 78, 95124 Catania, Italy; (S.L.); (R.S.); (G.A.M.); (C.B.); (B.G.); (G.S.); (A.S.); (A.C.); (G.M.)
| | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Bachireddy P, Azizi E, Burdziak C, Nguyen VN, Ennis CS, Maurer K, Park CY, Choo ZN, Li S, Gohil SH, Ruthen NG, Ge Z, Keskin DB, Cieri N, Livak KJ, Kim HT, Neuberg DS, Soiffer RJ, Ritz J, Alyea EP, Pe'er D, Wu CJ. Mapping the evolution of T cell states during response and resistance to adoptive cellular therapy. Cell Rep 2021; 37:109992. [PMID: 34758319 PMCID: PMC9035342 DOI: 10.1016/j.celrep.2021.109992] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 06/23/2021] [Accepted: 10/21/2021] [Indexed: 01/06/2023] Open
Abstract
To elucidate mechanisms by which T cells eliminate leukemia, we study donor lymphocyte infusion (DLI), an established immunotherapy for relapsed leukemia. We model T cell dynamics by integrating longitudinal, multimodal data from 94,517 bone marrow-derived single T cell transcriptomes in addition to chromatin accessibility and single T cell receptor sequencing from patients undergoing DLI. We find that responsive tumors are defined by enrichment of late-differentiated T cells before DLI and rapid, durable expansion of early differentiated T cells after treatment, highly similar to "terminal" and "precursor" exhausted subsets, respectively. Resistance, in contrast, is defined by heterogeneous T cell dysfunction. Surprisingly, early differentiated T cells in responders mainly originate from pre-existing and novel clonotypes recruited to the leukemic microenvironment, rather than the infusion. Our work provides a paradigm for analyzing longitudinal single-cell profiling of scenarios beyond adoptive cell therapy and introduces Symphony, a Bayesian approach to infer regulatory circuitry underlying T cell subsets, with broad relevance to exhaustion antagonists across cancers.
Collapse
Affiliation(s)
- Pavan Bachireddy
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02115, USA; Department of Hematopoietic Biology & Malignancy, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Cancer Prevention and Research Institute of Texas (CPRIT) Scholar in Cancer Research, Austin, TX 78701, USA.
| | - Elham Azizi
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Biomedical Engineering and Irving Institute for Cancer Dynamics, Columbia University, New York, NY 10027, USA.
| | - Cassandra Burdziak
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Tri-Institutional Training Program in Computational Biology and Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Vinhkhang N Nguyen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Christina S Ennis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Katie Maurer
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Cameron Y Park
- Department of Biomedical Engineering and Irving Institute for Cancer Dynamics, Columbia University, New York, NY 10027, USA
| | - Zi-Ning Choo
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Shuqiang Li
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Satyen H Gohil
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Neil G Ruthen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Zhongqi Ge
- Department of Hematopoietic Biology & Malignancy, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Derin B Keskin
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Nicoletta Cieri
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Kenneth J Livak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Haesook T Kim
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Donna S Neuberg
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Robert J Soiffer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Jerome Ritz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Edwin P Alyea
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Dana Pe'er
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute of Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Catherine J Wu
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
8
|
Poonsombudlert K, Kewcharoen J, Prueksapraopong C, Limpruttidham N. Prophylactic donor lymphocyte infusion for relapse prevention: a meta-analysis. Jpn J Clin Oncol 2020; 50:661-670. [PMID: 32100001 DOI: 10.1093/jjco/hyaa022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/28/2020] [Accepted: 01/31/2020] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVE Primary disease relapse (PDR) of malignant hematologic conditions after standard hematopoietic stem cell transplant (HSCT) is one of the most challenging diseases; therefore ongoing researches are aiming at relapse prevention and minimizing the transplant-related side effects. Prophylactic donor lymphocytes (pDLI) had been proposed as a valuable strategy for PDR prevention, but early studies had been discouraging due to the limited benefit and possible association with acute graft-versus-host disease (aGVHD). Therefore, we conducted a meta-analysis to evaluate the association between pDLI use, PDR, aGVHD and OS. METHOD We performed a comprehensive literature search in MEDLINE, Cochrane library and Embase database from inception to May 2019 for studies that evaluated the association between pDLI and PDR. We conducted a random effect meta-analysis of 9 studies involving a total of 748 participants (pDLI = 398, non-pDLI = 350) and reported the pooled odd ratio (OR) for association of pDLI use, PDR, aGVHD and OS. RESULT We found a significant decreased odd of PDR in the pDLI group (pooled OR = 0.42, 95% CI 0.30-0.58, I2 = 0%), but there was no significant increased odd of aGVHD (pooled OR of 0.98, 95% CI 0.56-1.72, I2 = 0.8%). We also found that there was an increased odd of overall survival (OS) (pooled OR 3.17, 95% CI 1.85-5.45, I2 = 50.2%). CONCLUSION There are significantly decreased odd of PDR and increased odd of OS in the pDLI group compared to the control group, but there is no statistically significant increased odd of aGVHD as suggested by previous studies. We concluded that pDLI is a potentially valuable method for post-transplant PDR prevention.
Collapse
Affiliation(s)
| | - Jakrin Kewcharoen
- University of Hawaii, Internal Medicine Residency Program, Honolulu, HI, USA, and
| | | | - Nath Limpruttidham
- University of Hawaii, Internal Medicine Residency Program, Honolulu, HI, USA, and
| |
Collapse
|
9
|
Intra-bone donor lymphocyte infusion at relapse: clinical outcome is associated with presence of CD8+ cells in the marrow. Bone Marrow Transplant 2019; 55:974-978. [PMID: 31455896 PMCID: PMC7198473 DOI: 10.1038/s41409-019-0632-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 06/04/2019] [Accepted: 06/20/2019] [Indexed: 11/08/2022]
|
10
|
Wu X, Liu Q. Prophylaxis and treatment of relapse after haploidentical stem cell transplantation: What is known vs unknown? Semin Hematol 2019; 56:209-214. [PMID: 31202432 DOI: 10.1053/j.seminhematol.2019.04.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 04/10/2019] [Indexed: 11/11/2022]
Abstract
In recent years, the human leukocyte antigen-haploidentical stem cell transplantation (haplo-SCT) approach is an attractive option for patients who require transplantation, but relapse is still the main reason that affects the curative effect of transplantation. Some studies have shown that haplo-SCT is superior to sibling or unrelated matching donor transplantation in preventing leukemia relapse after transplantation. In this review, we discussed the known and unknown aspects of relapse post haplo-SCT. Encouragingly, haplo-SCT experienced lower or similar incidence of relapse. But there is currently a lack of multicenter prospective studies evaluating the outcomes of different haplo-SCT strategies. The combination of common prophylactic strategies and pre-emptive interventions might help prevent relapse after transplantation. Novel methods such as target drugs therapy and chimeric antigen receptor T cell therapy may be useful in treatment of relapse.
Collapse
Affiliation(s)
- Xiuli Wu
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
11
|
Ma H, Padmanabhan Iyer S, Parmar S, Gong Y. Adoptive cell therapy for acute myeloid leukemia. Leuk Lymphoma 2019; 60:1370-1380. [PMID: 30628504 DOI: 10.1080/10428194.2018.1553300] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Hongbing Ma
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | | | - Simrit Parmar
- Department of Lymphoma & Myeloma, MD Anderson Cancer Center, Texas University, Houston, TX, USA
| | - Yuping Gong
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
12
|
A retrospective comparison of allogenic and autologous chimeric antigen receptor T cell therapy targeting CD19 in patients with relapsed/refractory acute lymphoblastic leukemia. Bone Marrow Transplant 2018; 54:1208-1217. [PMID: 30518980 DOI: 10.1038/s41409-018-0403-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 10/30/2018] [Accepted: 11/02/2018] [Indexed: 12/31/2022]
Abstract
The source of CAR T cells can be autologous (autoCAR) or allogeneic (alloCAR). The latter is seen in patients with a history of allogeneic hematopoietic stem cell transplantation, and can be either donor-derived (DD-alloCAR) or recipient-derived (RD-alloCAR). While autoCAR is activated by CAR only, alloCAR receives activation signals from both T-cell receptor (TCR) and CAR. As a result, the biological differences could impact clinical outcomes. We retrospectively reviewed 31 patients: 17 received autoCAR, 11 received RD-alloCAR, and 3 received DD-alloCAR. After a median follow-up of 9 months, CR rate was 88.2% (95% CI 63.6-98.5%) in autoCAR and 100% (95% CI 71.5-100%) in RD-alloCAR. The median peak expansion in the autoCAR was significantly higher than the RD-alloCAR group (p = 0.007). RD-alloCAR group had significantly less patients with severe CRS (Grade ≥ 3) than the autoCAR group (p = 0.049). Acute graft-versus-host disease (GVHD) occurred in 2 (18.2%) of RD-alloCAR patients and 1 (33.3%) of DD-alloCAR patients. Univariate subgroup analysis of alloCAR group showed the presence of cGVHD at the time of T-cell collection was significantly associated with less than 6-month relapses (p = 0.022). RD-alloCAR patients with or without cGVHD at PBMC collection did not differ regarding the peak CAR T-cell expansion, CRS grades and OS.
Collapse
|
13
|
Abstract
A number of modalities including both pharmaceutical and cell-based treatments have long been tested and developed to prevent and treat relapses after allogeneic stem cell transplantation (allo-HSCT). The ability of donor T cells to recognize antigenic structures on leukemic cell surfaces and destroy them is a well-known fact. Based on this fact, the idea of using donor T cells to contribute to the development of adoptive immunotherapy has emerged. Donor lymphocytes are easy to obtain and donor lymphocyte infusions (DLI) have a simple rational while this treatment modality is an effective example of cellular therapy. The group of chronic myeloid leukemia patients who are more likely to benefit from DLI include: a) patients in the chronic phase of hematologic relapse and b) patients with molecular/cytogenetic relapse. DLI appear to be an appropriate treatment option to be used in combination with conventional chemotherapy or hypomethylating agents in the treatment of post-allo-HSCT relapse for acute myeloid leukemia and myelodysplastic syndrome, if:) the burden of tumor is low b) the relapse is at a molecular level rather than an overt hematologic relapse c) the patient has favorable cytogenetic characteristics d) time interval between transplantation and relapse is relatively longer (>5 months) e) response could be obtained after salvage therapies. In the event that minimal residual disease (MRD) or increasing mixed chimerism is detected, prompt administration of DLI for prophylactic purposes without waiting for a manifest relapse, was found to be effective in inducing a full donor chimerism and overcoming MRD and eventually preventing a manifest relapse.
Collapse
Affiliation(s)
- Selami Koçak Toprak
- Ankara University, School of Medicine, Department of Hematology, Cebeci Hospital, Dikimevi, Ankara, Turkey.
| |
Collapse
|
14
|
Liu L, Chang YJ, Xu LP, Zhang XH, Wang Y, Liu KY, Huang XJ. Reversal of T Cell Exhaustion by the First Donor Lymphocyte Infusion Is Associated with the Persistently Effective Antileukemic Responses in Patients with Relapsed AML after Allo-HSCT. Biol Blood Marrow Transplant 2018; 24:1350-1359. [PMID: 29649617 DOI: 10.1016/j.bbmt.2018.03.030] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 03/29/2018] [Indexed: 01/28/2023]
Abstract
Donor lymphocyte infusion (DLI) is an effective approach to treat acute myelogenous leukemia (AML) relapse after allogeneic hematopoietic stem cell transplantation (allo-HSCT) that significantly improves the survival of relapsed patients. However, the mechanism of an effective antileukemic response following DLI in AML relapse remains elusive. Here, we investigated the role of T cell exhaustion in AML relapse after allo-HSCT in prospective cohorts of 41 patients with the first AML relapse and 41 nonrelapsed AML control subjects after allo-HSCT and determined whether DLI exerts effective antileukemic effects by reversing T cell exhaustion in the relapsed cohorts by detecting the phenotypes and functions of T cells using flow cytometry. We found that both CD4+ and CD8+ T cells experienced exhaustion with upregulated coexpression of PD-1 and Tim-3, and functional impairments in cytokine production, proliferation, and cytotoxic potentials. The reversal of T cell exhaustion by the first DLI is associated with persistent complete remission in relapsed AML patients. In addition, the reversal of T cell-exhausted status after successful DLI in bone marrow was concurrent with the mitigated inversion of CD4/CD8 T cell ratio. In conclusion, our study shows a clinical correlation between T cell exhaustion and AML relapse after allo-HSCT, and uncovers the role of reversing T cell exhaustion in the antileukemic response by DLI and identifies possible immunological markers to evaluate and predict the graft-versus-leukemia effects induced by DLI.
Collapse
Affiliation(s)
- Long Liu
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital & Peking University Institute of Hematology, Beijing, China
| | - Ying-Jun Chang
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital & Peking University Institute of Hematology, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Lan-Ping Xu
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital & Peking University Institute of Hematology, Beijing, China
| | - Xiao-Hui Zhang
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital & Peking University Institute of Hematology, Beijing, China
| | - Yu Wang
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital & Peking University Institute of Hematology, Beijing, China
| | - Kai-Yan Liu
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital & Peking University Institute of Hematology, Beijing, China
| | - Xiao-Jun Huang
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital & Peking University Institute of Hematology, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.
| |
Collapse
|
15
|
Xuan L, Fan Z, Zhang Y, Zhou H, Huang F, Dai M, Nie D, Lin D, Xu N, Guo X, Jiang Q, Sun J, Xiao Y, Liu Q. Sequential intensified conditioning followed by prophylactic DLI could reduce relapse of refractory acute leukemia after allo-HSCT. Oncotarget 2018; 7:32579-91. [PMID: 27081039 PMCID: PMC5078035 DOI: 10.18632/oncotarget.8691] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 03/28/2016] [Indexed: 01/25/2023] Open
Abstract
The major obstacle is leukemia relapse for refractory leukemia undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT). We previously introduced a strategy of sequential intensified conditioning and early rapid immunosupressant withdrawal for refractory leukemia undergoing allo-HSCT, with 5-year overall survival (OS) and 3-year relapse rate of 44.6% and 33.3%. To reduce leukemia relapse, prophylactic donor lymphocyte infusion (DLI) was administered based on our historical strategy. A total of 153 refractory advanced acute leukemia patients were enrolled in this prospective study. According to the availability of donor lymphocytes and the criteria for DLI, 144 patients surviving day +60 were divided into two groups (80 DLI versus 64 non-DLI). The relapse rate was less and OS was better in patients receiving DLI than in those not receiving DLI (22.7% vs 33.9%, P=0.048; 58.1% vs 54.9%, P=0.043). The non-relapse mortality (NRM) was similar between DLI and non-DLI groups (P=0.104). Overall, the 5-year overall and disease-free survival post-transplantation were 51.1%±5.7% and 49.2%±5.3%. The 5-year relapse rate and NRM were 27.3%±4.4% and 29.7%±5.3%. Multivariate analysis revealed that lower bone marrow blasts on day 0, DLI and chronic graft-versus-host disease were associated with less relapse and better OS. The strategy of sequential intensified conditioning followed by early immunosupressant withdrawal and DLI could reduce relapse of refractory acute leukemia after allo-HSCT and improve survival.
Collapse
Affiliation(s)
- Li Xuan
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhiping Fan
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yu Zhang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Hongsheng Zhou
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Fen Huang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Min Dai
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Danian Nie
- Department of Hematology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Dongjun Lin
- Department of Hematology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Na Xu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xutao Guo
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qianli Jiang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jing Sun
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yang Xiao
- Department of Hematology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou 510010, China
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
16
|
Ueha S, Yokochi S, Ishiwata Y, Kosugi-Kanaya M, Shono Y, Shibayama S, Ito S, Matsushima K. Combination of anti-CD4 antibody treatment and donor lymphocyte infusion ameliorates graft-versus-host disease while preserving graft-versus-tumor effects in murine allogeneic hematopoietic stem cell transplantation. Cancer Sci 2017; 108:1967-1973. [PMID: 28787768 PMCID: PMC5623734 DOI: 10.1111/cas.13346] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 07/25/2017] [Accepted: 07/27/2017] [Indexed: 11/29/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo‐HSCT) is not only a well‐established immunotherapy for hematologic malignancies, but is potentially useful for treating solid tumors refractory to available therapies. However, application of allo‐HSCT to solid tumors is limited, despite the beneficial antitumor effects, by the risk of graft‐versus‐host disease (GVHD). CD4+ T cells have been implicated in several aspects of GVHD, and suppress antitumor CD8+ T‐cell responses. In the present study, we investigated clinically applicable allo‐HSCT protocols designed to maximize antitumor effects while reducing the risk of GVHD. We used a mouse model of allo‐HSCT with s.c. tumors. We found that myeloablative conditioning was associated with better inhibition of tumor growth but with severe acute GVHD. Early treatment with anti‐CD4 mAb substantially ameliorated GVHD while preserving antitumor effects, leading to improved survival in myeloablative allo‐HSCT. Late treatment with anti‐CD4 mAb also ameliorated GVHD to some extent. Donor lymphocyte infusion in GVHD mice treated with anti‐CD4 mAb further suppressed tumor growth without exacerbating GVHD. Collectively, our results suggest that myeloablative allo‐HSCT followed by anti‐CD4 mAb treatment and donor lymphocyte infusion could be a potent and safe immunotherapy for patients with cancers refractory to available therapies.
Collapse
Affiliation(s)
- Satoshi Ueha
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shoji Yokochi
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,IDAC Theranostics Inc., Tokyo, Japan
| | - Yoshiro Ishiwata
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,IDAC Theranostics Inc., Tokyo, Japan
| | - Mizuha Kosugi-Kanaya
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Hematology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Yusuke Shono
- Department of Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Shiro Shibayama
- Research Center of Immunology, Tsukuba Institute, ONO Pharmaceutical Co., Ltd., Tsukuba, Japan
| | - Satoru Ito
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,IDAC Theranostics Inc., Tokyo, Japan
| | - Kouji Matsushima
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
17
|
Abstract
Historically, immune-based therapies have played a leading role in the treatment of hematologic malignancies, with the efficacy of stem cell transplantation largely attributable to donor immunity against malignant cells. As new and more targeted immunotherapies have developed, their role in the treatment of hematologic malignancies is evolving and expanding. Herein, we discuss approaches for antigen discovery and review known and novel tumor antigens in hematologic malignancies. We further explore the role of established and investigational immunotherapies in hematologic malignancies, with a focus on personalization of treatment modalities such as cancer vaccines and adoptive cell therapy. Finally, we identify areas of active investigation and development. Immunotherapy is at an exciting crossroads for the treatment of hematologic malignancies, with further investigation aimed at producing effective, targeted immune therapies that maximize antitumor effects while minimizing toxicity.
Collapse
Affiliation(s)
- David A. Braun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts, USA
| | - Catherine J. Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
18
|
Tosolini M, Pont F, Poupot M, Vergez F, Nicolau-Travers ML, Vermijlen D, Sarry JE, Dieli F, Fournié JJ. Assessment of tumor-infiltrating TCRV γ9V δ2 γδ lymphocyte abundance by deconvolution of human cancers microarrays. Oncoimmunology 2017; 6:e1284723. [PMID: 28405516 DOI: 10.1080/2162402x.2017.1284723] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/12/2017] [Accepted: 01/13/2017] [Indexed: 10/20/2022] Open
Abstract
Most human blood γδ cells are cytolytic TCRVγ9Vδ2+ lymphocytes with antitumor activity. They are currently investigated in several clinical trials of cancer immunotherapy but so far, their tumor infiltration has not been systematically explored across human cancers. Novel algorithms allowing the deconvolution of bulk tumor transcriptomes to find the relative proportions of infiltrating leucocytes, such as CIBERSORT, should be appropriate for this aim but in practice they fail to accurately recognize γδ T lymphocytes. Here, by implementing machine learning from microarray data, we first improved the computational identification of blood-derived TCRVγ9Vδ2+ γδ lymphocytes and then applied this strategy to assess their abundance as tumor infiltrating lymphocytes (γδ TIL) in ∼10,000 cancer biopsies from 50 types of hematological and solid malignancies. We observed considerable inter-individual variation of TCRVγ9Vδ2+γδ TIL abundance both within each type and across the spectrum of cancers tested. We report their prominence in B cell-acute lymphoblastic leukemia (B-ALL), acute promyelocytic leukemia (M3-AML) and chronic myeloid leukemia (CML) as well as in inflammatory breast, prostate, esophagus, pancreas and lung carcinoma. Across all cancers, the abundance of αβ TILs and TCRVγ9Vδ2+ γδ TILs did not correlate. αβ TIL abundance paralleled the mutational load of tumors and positively correlated with inflammation, infiltration of monocytes, macrophages and dendritic cells (DC), antigen processing and presentation, and cytolytic activity, in line with an association with a favorable outcome. In contrast, the abundance of TCRVγ9Vδ2+ γδ TILs did not correlate with these hallmarks and was variably associated with outcome, suggesting that distinct contexts underlie TCRVγ9Vδ2+ γδ TIL and αβ TIL mobilizations in cancer.
Collapse
Affiliation(s)
- Marie Tosolini
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France; INSERM U1037-Université Paul Sabatier-CNRS ERL5294, Université de Toulouse, Toulouse, France; Laboratoire d'Excellence TOUCAN, Toulouse, France; Programme Hospitalo-Universitaire en Cancérologie CAPTOR, Toulouse, France; Pôle Technologique du Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France; Institut Universitaire du Cancer de Toulouse (IUCT), Toulouse, France
| | - Frédéric Pont
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France; INSERM U1037-Université Paul Sabatier-CNRS ERL5294, Université de Toulouse, Toulouse, France; Pôle Technologique du Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France
| | - Mary Poupot
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France; INSERM U1037-Université Paul Sabatier-CNRS ERL5294, Université de Toulouse, Toulouse, France; Laboratoire d'Excellence TOUCAN, Toulouse, France; Programme Hospitalo-Universitaire en Cancérologie CAPTOR, Toulouse, France
| | - François Vergez
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France; INSERM U1037-Université Paul Sabatier-CNRS ERL5294, Université de Toulouse, Toulouse, France; Institut Universitaire du Cancer de Toulouse (IUCT), Toulouse, France
| | | | - David Vermijlen
- Central Laboratory for Advanced Diagnostics and Biomedical Research (CLADIBIOR), University of Palermo , Palermo, Italy
| | - Jean-Emmanuel Sarry
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France; INSERM U1037-Université Paul Sabatier-CNRS ERL5294, Université de Toulouse, Toulouse, France; Programme Hospitalo-Universitaire en Cancérologie CAPTOR, Toulouse, France
| | - Francesco Dieli
- Department of Biopharmacy - Institute for Medical Immunology (IMI), Université Libre de Bruxelles , Bruxelles, Belgium
| | - Jean-Jacques Fournié
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France; INSERM U1037-Université Paul Sabatier-CNRS ERL5294, Université de Toulouse, Toulouse, France; Laboratoire d'Excellence TOUCAN, Toulouse, France; Programme Hospitalo-Universitaire en Cancérologie CAPTOR, Toulouse, France
| |
Collapse
|
19
|
Castagna L, Sarina B, Bramanti S, Perseghin P, Mariotti J, Morabito L. Donor lymphocyte infusion after allogeneic stem cell transplantation. Transfus Apher Sci 2016; 54:345-55. [PMID: 27216544 DOI: 10.1016/j.transci.2016.05.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Allogeneic stem cell transplantation (allo-SCT) is considered the cornerstone in the treatment of several malignant and not malignant hematological diseases. However, relapse of hematological disease after allo-SCT is considered the most challenging point in the field. The risk can be reduced through optimal patients, donor and disease selection before allo-SCT, but harnessing donor immune system is an appealing way to treat or avoid disease relapse. Donor lymphocyte infusion (DLI) is a simple and effective therapy after allo-SCT. In this paper, the efficacy of DLI will be analyzed in different hematological diseases, focusing also on their therapeutic or pre-emptive use.
Collapse
Affiliation(s)
- Luca Castagna
- Programma Trapianto Humanitas Cancer Center, Istituto Clinico Humanitas, Rozzano, Italy.
| | - Barbara Sarina
- Programma Trapianto Humanitas Cancer Center, Istituto Clinico Humanitas, Rozzano, Italy
| | - Stefania Bramanti
- Programma Trapianto Humanitas Cancer Center, Istituto Clinico Humanitas, Rozzano, Italy
| | | | - Jacopo Mariotti
- Programma Trapianto Humanitas Cancer Center, Istituto Clinico Humanitas, Rozzano, Italy
| | - Lucio Morabito
- Programma Trapianto Humanitas Cancer Center, Istituto Clinico Humanitas, Rozzano, Italy
| |
Collapse
|