251
|
Adoptive T-cell therapy: adverse events and safety switches. Clin Transl Immunology 2014; 3:e17. [PMID: 25505965 PMCID: PMC4232067 DOI: 10.1038/cti.2014.11] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 05/08/2014] [Accepted: 05/08/2014] [Indexed: 12/17/2022] Open
Abstract
The potential of adoptive T-cell therapy in effecting complete and durable responses has been demonstrated in a number of malignant and infectious diseases. Ongoing progress in T-cell engineering has given cause for optimism in the broader clinical applicability of this approach. However, the development of more potent T cells is checked by safety concerns, highlighted by the occurrence of on-target and off-target toxicities that, although uncommon, have been fatal on occasions. Timely pharmacological intervention is effective in the management of a majority of adverse events but adoptively transferred T cells can persist long term, along with any unwanted effects. A recently validated cellular safety switch, inducible caspase 9 (iCasp9), has the potential to mitigate the risks of T-cell therapy by enabling the elimination of transferred T cells if required. In haematopoietic stem cell transplantation, iCasp9-modified donor T cells can be rapidly eliminated in the event of graft-versus-host disease. This review presents an overview of the risks associated with modern T-cell therapy and the development, clinical results and potential future application of the iCasp9 safety switch.
Collapse
|
252
|
Abstract
Proof-of-concept studies have demonstrated the therapeutic potential of engineered T cells. Transfer of recombinant antigen-specific T cell receptors (TCR) and chimaeric antigen receptors (CARs) against tumour and viral antigens are under investigation by multiple approaches, including viral- and nonviral-mediated gene transfer into both autologous and allogeneic T cell populations. There have been notable successes recently using viral vector-mediated transfer of CARs specific for B cell antigens, but also reports of anticipated and unanticipated complications in these and other studies. We review progress in this promising area of cellular therapy, and consider developments in antigen receptor therapies including the application of emerging gene-editing technologies.
Collapse
Affiliation(s)
- Waseem Qasim
- Molecular & Cellular Immunology, Institute of Child Health, University College London, London, UK; Great Ormond Street Hospital Trust, London, UK
| | | |
Collapse
|
253
|
Lepore M, de Lalla C, Gundimeda SR, Gsellinger H, Consonni M, Garavaglia C, Sansano S, Piccolo F, Scelfo A, Häussinger D, Montagna D, Locatelli F, Bonini C, Bondanza A, Forcina A, Li Z, Ni G, Ciceri F, Jenö P, Xia C, Mori L, Dellabona P, Casorati G, De Libero G. A novel self-lipid antigen targets human T cells against CD1c(+) leukemias. ACTA ACUST UNITED AC 2014; 211:1363-77. [PMID: 24935257 PMCID: PMC4076585 DOI: 10.1084/jem.20140410] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
T cells that recognize self-lipids presented by CD1c are frequent in the peripheral blood of healthy individuals and kill transformed hematopoietic cells, but little is known about their antigen specificity and potential antileukemia effects. We report that CD1c self-reactive T cells recognize a novel class of self-lipids, identified as methyl-lysophosphatidic acids (mLPAs), which are accumulated in leukemia cells. Primary acute myeloid and B cell acute leukemia blasts express CD1 molecules. mLPA-specific T cells efficiently kill CD1c(+) acute leukemia cells, poorly recognize nontransformed CD1c-expressing cells, and protect immunodeficient mice against CD1c(+) human leukemia cells. The identification of immunogenic self-lipid antigens accumulated in leukemia cells and the observed leukemia control by lipid-specific T cells in vivo provide a new conceptual framework for leukemia immune surveillance and possible immunotherapy.
Collapse
Affiliation(s)
- Marco Lepore
- Experimental Immunology, Department of Biomedicine, University Hospital Basel; Nuclear Magnetic Resonance Laboratory, Department of Chemistry; and Department of Biochemistry, Biozentrum; University of Basel, 4056 Basel, Switzerland Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Claudia de Lalla
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - S Ramanjaneyulu Gundimeda
- Experimental Immunology, Department of Biomedicine, University Hospital Basel; Nuclear Magnetic Resonance Laboratory, Department of Chemistry; and Department of Biochemistry, Biozentrum; University of Basel, 4056 Basel, Switzerland
| | - Heiko Gsellinger
- Experimental Immunology, Department of Biomedicine, University Hospital Basel; Nuclear Magnetic Resonance Laboratory, Department of Chemistry; and Department of Biochemistry, Biozentrum; University of Basel, 4056 Basel, Switzerland
| | - Michela Consonni
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Claudio Garavaglia
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Sebastiano Sansano
- Experimental Immunology, Department of Biomedicine, University Hospital Basel; Nuclear Magnetic Resonance Laboratory, Department of Chemistry; and Department of Biochemistry, Biozentrum; University of Basel, 4056 Basel, Switzerland
| | - Francesco Piccolo
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Andrea Scelfo
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Daniel Häussinger
- Experimental Immunology, Department of Biomedicine, University Hospital Basel; Nuclear Magnetic Resonance Laboratory, Department of Chemistry; and Department of Biochemistry, Biozentrum; University of Basel, 4056 Basel, Switzerland
| | - Daniela Montagna
- Laboratorio di Immunologia, Dipartimento di Pediatria, Università di Pavia and Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology-Oncology, IRCCS Bambino Gesù Hospital, 00165 Rome, Italy
| | - Chiara Bonini
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Attilio Bondanza
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Alessandra Forcina
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Zhiyuan Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Guanghui Ni
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Fabio Ciceri
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Paul Jenö
- Experimental Immunology, Department of Biomedicine, University Hospital Basel; Nuclear Magnetic Resonance Laboratory, Department of Chemistry; and Department of Biochemistry, Biozentrum; University of Basel, 4056 Basel, Switzerland
| | - Chengfeng Xia
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Lucia Mori
- Experimental Immunology, Department of Biomedicine, University Hospital Basel; Nuclear Magnetic Resonance Laboratory, Department of Chemistry; and Department of Biochemistry, Biozentrum; University of Basel, 4056 Basel, Switzerland Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research, Singapore 138648
| | - Paolo Dellabona
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Giulia Casorati
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Gennaro De Libero
- Experimental Immunology, Department of Biomedicine, University Hospital Basel; Nuclear Magnetic Resonance Laboratory, Department of Chemistry; and Department of Biochemistry, Biozentrum; University of Basel, 4056 Basel, Switzerland Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research, Singapore 138648
| |
Collapse
|
254
|
Peccatori J, Forcina A, Clerici D, Crocchiolo R, Vago L, Stanghellini MTL, Noviello M, Messina C, Crotta A, Assanelli A, Marktel S, Olek S, Mastaglio S, Giglio F, Crucitti L, Lorusso A, Guggiari E, Lunghi F, Carrabba M, Tassara M, Battaglia M, Ferraro A, Carbone MR, Oliveira G, Roncarolo MG, Rossini S, Bernardi M, Corti C, Marcatti M, Patriarca F, Zecca M, Locatelli F, Bordignon C, Fleischhauer K, Bondanza A, Bonini C, Ciceri F. Sirolimus-based graft-versus-host disease prophylaxis promotes the in vivo expansion of regulatory T cells and permits peripheral blood stem cell transplantation from haploidentical donors. Leukemia 2014; 29:396-405. [PMID: 24897508 DOI: 10.1038/leu.2014.180] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 04/14/2014] [Accepted: 05/19/2014] [Indexed: 01/05/2023]
Abstract
Hematopoietic stem cell transplantation (HSCT) from human leukocyte antigen (HLA) haploidentical family donors is a promising therapeutic option for high-risk hematologic malignancies. Here we explored in 121 patients, mostly with advanced stage diseases, a sirolimus-based, calcineurin-inhibitor-free prophylaxis of graft-versus-host disease (GvHD) to allow the infusion of unmanipulated peripheral blood stem cell (PBSC) grafts from partially HLA-matched family donors (TrRaMM study, Eudract 2007-5477-54). Conditioning regimen was based on treosulfan and fludarabine, and GvHD prophylaxis on antithymocyte globulin Fresenius (ATG-F), rituximab and oral administration of sirolimus and mycophenolate. Neutrophil and platelet engraftment occurred in median at 17 and 19 days after HSCT, respectively, and full donor chimerism was documented in patients' bone marrow since the first post-transplant evaluation. T-cell immune reconstitution was rapid, and high frequencies of circulating functional T-regulatory cells (Treg) were documented during sirolimus prophylaxis. Incidence of acute GvHD grade II-IV was 35%, and occurrence and severity correlated negatively with Treg frequency. Chronic GvHD incidence was 47%. At 3 years after HSCT, transpant-related mortality was 31%, relapse incidence 48% and overall survival 25%. In conclusion, GvHD prophylaxis with sirolimus-mycophenolate-ATG-F-rituximab promotes a rapid immune reconstitution skewed toward Tregs, allowing the infusion of unmanipulated haploidentical PBSC grafts.
Collapse
Affiliation(s)
- J Peccatori
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Gene Therapy and Stem Cells, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - A Forcina
- 1] Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Gene Therapy and Stem Cells, IRCCS San Raffaele Scientific Institute, Milan, Italy [2] Experimental Hematology Unit, Division of Immunology, Infectious Diseases and Transplants, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - D Clerici
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Gene Therapy and Stem Cells, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - R Crocchiolo
- 1] Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Gene Therapy and Stem Cells, IRCCS San Raffaele Scientific Institute, Milan, Italy [2] Humanitas Cancer Center, Rozzano, Italy
| | - L Vago
- 1] Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Gene Therapy and Stem Cells, IRCCS San Raffaele Scientific Institute, Milan, Italy [2] Unit of Molecular and Functional Immunogenetics, Division of Regenerative Medicine, Gene Therapy and Stem Cells, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - M T L Stanghellini
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Gene Therapy and Stem Cells, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - M Noviello
- Experimental Hematology Unit, Division of Immunology, Infectious Diseases and Transplants, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - C Messina
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Gene Therapy and Stem Cells, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - A Crotta
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Gene Therapy and Stem Cells, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - A Assanelli
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Gene Therapy and Stem Cells, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - S Marktel
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Gene Therapy and Stem Cells, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - S Olek
- Epiontis GmbH, Berlin, Germany
| | - S Mastaglio
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Gene Therapy and Stem Cells, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - F Giglio
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Gene Therapy and Stem Cells, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - L Crucitti
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Gene Therapy and Stem Cells, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - A Lorusso
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Gene Therapy and Stem Cells, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - E Guggiari
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Gene Therapy and Stem Cells, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - F Lunghi
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Gene Therapy and Stem Cells, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - M Carrabba
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Gene Therapy and Stem Cells, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - M Tassara
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Gene Therapy and Stem Cells, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - M Battaglia
- San Raffaele Diabetes Research Institute, Division of Immunology, Infectious Diseases and Transplants, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - A Ferraro
- San Raffaele Diabetes Research Institute, Division of Immunology, Infectious Diseases and Transplants, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - M R Carbone
- Experimental Hematology Unit, Division of Immunology, Infectious Diseases and Transplants, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - G Oliveira
- Experimental Hematology Unit, Division of Immunology, Infectious Diseases and Transplants, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - M G Roncarolo
- 1] Pediatric Immunology, Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Gene Therapy and Stem Cells, IRCCS San Raffaele Scientific Institute, Milan, Italy [2] San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), Division of Regenerative Medicine, Gene Therapy and Stem Cells, San Raffaele Scientific Institute, Milan, Italy [3] 'Vita-Salute' San Raffaele University, Milan, Italy
| | - S Rossini
- Immunohematology and Transfusion Medicine Service, Division of Regenerative Medicine, Gene Therapy and Stem Cells, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - M Bernardi
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Gene Therapy and Stem Cells, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - C Corti
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Gene Therapy and Stem Cells, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - M Marcatti
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Gene Therapy and Stem Cells, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - F Patriarca
- Clinica Ematologica, Policlinico Universitario, Udine, Italy
| | - M Zecca
- Policlinico San Matteo, Pavia, Italy
| | | | - C Bordignon
- 1] 'Vita-Salute' San Raffaele University, Milan, Italy [2] MolMed SpA, Milan, Italy
| | - K Fleischhauer
- Unit of Molecular and Functional Immunogenetics, Division of Regenerative Medicine, Gene Therapy and Stem Cells, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - A Bondanza
- 1] Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Gene Therapy and Stem Cells, IRCCS San Raffaele Scientific Institute, Milan, Italy [2] Leukemia Immunotherapy Group, Division of Regenerative Medicine, Gene Therapy and Stem Cells, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - C Bonini
- Experimental Hematology Unit, Division of Immunology, Infectious Diseases and Transplants, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - F Ciceri
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Gene Therapy and Stem Cells, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
255
|
Castagna L, Crocchiolo R, Furst S, Bramanti S, El Cheikh J, Sarina B, Granata A, Mauro E, Faucher C, Mohty B, Harbi S, Chabannon C, Carlo-Stella C, Santoro A, Blaise D. Bone Marrow Compared with Peripheral Blood Stem Cells for Haploidentical Transplantation with a Nonmyeloablative Conditioning Regimen and Post-transplantation Cyclophosphamide. Biol Blood Marrow Transplant 2014; 20:724-9. [DOI: 10.1016/j.bbmt.2014.02.001] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 02/04/2014] [Indexed: 12/01/2022]
|
256
|
Long-term outcome after haploidentical stem cell transplant and infusion of T cells expressing the inducible caspase 9 safety transgene. Blood 2014; 123:3895-905. [PMID: 24753538 DOI: 10.1182/blood-2014-01-551671] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Adoptive transfer of donor-derived T lymphocytes expressing a safety switch may promote immune reconstitution in patients undergoing haploidentical hematopoietic stem cell transplant (haplo-HSCT) without the risk for uncontrolled graft versus host disease (GvHD). Thus, patients who develop GvHD after infusion of allodepleted donor-derived T cells expressing an inducible human caspase 9 (iC9) had their disease effectively controlled by a single administration of a small-molecule drug (AP1903) that dimerizes and activates the iC9 transgene. We now report the long-term follow-up of 10 patients infused with such safety switch-modified T cells. We find long-term persistence of iC9-modified (iC9-T) T cells in vivo in the absence of emerging oligoclonality and a robust immunologic benefit, mediated initially by the infused cells themselves and subsequently by an apparently accelerated reconstitution of endogenous naive T lymphocytes. As a consequence, these patients have immediate and sustained protection from major pathogens, including cytomegalovirus, adenovirus, BK virus, and Epstein-Barr virus in the absence of acute or chronic GvHD, supporting the beneficial effects of this approach to immune reconstitution after haplo-HSCT. This study was registered at www.clinicaltrials.gov as #NCT00710892.
Collapse
|
257
|
Ramírez N, Beloki L, Ciaúrriz M, Rodríguez-Calvillo M, Escors D, Mansilla C, Bandrés E, Olavarría E. Impact of T cell selection methods in the success of clinical adoptive immunotherapy. Cell Mol Life Sci 2014; 71:1211-24. [PMID: 24077876 PMCID: PMC11113470 DOI: 10.1007/s00018-013-1463-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 08/20/2013] [Accepted: 08/23/2013] [Indexed: 12/11/2022]
Abstract
Chemotherapy and/or radiotherapy regular regimens used for conditioning of recipients of hematopoietic stem cell transplantation (SCT) induce a period of transient profound immunosuppression. The onset of a competent immunological response, such as the appearance of viral-specific T cells, is associated with a lower incidence of viral infections after haematopoietic transplantation. The rapid development of immunodominant peptide virus screening together with advances in the design of genetic and non-genetic viral- and tumoural-specific cellular selection strategies have opened new strategies for cellular immunotherapy in oncologic recipients who are highly sensitive to viral infections. However, the rapid development of cellular immunotherapy in SCT has disclosed the role of the T cell selection method in the modulation of functional cell activity and of in vivo secondary effects triggered following immunotherapy.
Collapse
Affiliation(s)
- Natalia Ramírez
- Oncohematology Research Group, Navarrabiomed, Miguel Servet Foundation, Irunlarrea 3 Street, 31008 Pamplona, Navarre Spain
| | - Lorea Beloki
- Oncohematology Research Group, Navarrabiomed, Miguel Servet Foundation, Irunlarrea 3 Street, 31008 Pamplona, Navarre Spain
| | - Miriam Ciaúrriz
- Oncohematology Research Group, Navarrabiomed, Miguel Servet Foundation, Irunlarrea 3 Street, 31008 Pamplona, Navarre Spain
| | - Mercedes Rodríguez-Calvillo
- Department of Haematology, Complejo Hospitalario de Navarra, Navarra Health Service, Irunlarrea 3 Street, 31008 Pamplona, Navarre Spain
| | - David Escors
- Immunomodulation Research Group, Navarrabiomed, Miguel Servet Foundation, Pamplona, Navarre Spain
| | - Cristina Mansilla
- Oncohematology Research Group, Navarrabiomed, Miguel Servet Foundation, Irunlarrea 3 Street, 31008 Pamplona, Navarre Spain
| | - Eva Bandrés
- Immunology Unit, Complejo Hospitalario de Navarra, Navarra Health Service, Pamplona, Spain
| | - Eduardo Olavarría
- Oncohematology Research Group, Navarrabiomed, Miguel Servet Foundation, Irunlarrea 3 Street, 31008 Pamplona, Navarre Spain
- Department of Haematology, Complejo Hospitalario de Navarra, Navarra Health Service, Irunlarrea 3 Street, 31008 Pamplona, Navarre Spain
| |
Collapse
|
258
|
Accelerating immune reconstitution after hematopoietic stem cell transplantation. Clin Transl Immunology 2014; 3:e11. [PMID: 25505959 PMCID: PMC4232061 DOI: 10.1038/cti.2014.2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 01/16/2014] [Accepted: 01/16/2014] [Indexed: 01/14/2023] Open
Abstract
Viral infections remain a significant cause of morbidity and mortality after hematopoietic stem cell transplantation. Pharmacologic agents are effective against some pathogens, but they are costly and can be associated with significant toxicities. Thus, many groups have investigated adoptive T-cell transfer as a means of hastening immune reconstitution and preventing and treating viral infections. This review discusses the immunotherapeutic strategies that have been explored.
Collapse
|
259
|
Abstract
Today human leukocyte antigen-haploidentical transplantation is a feasible option for patients with high-risk acute leukemia who do not have matched donors. Whether it is T-cell replete or T-cell depleted, it is still, however, associated with issues of transplant-related mortality and posttransplant leukemia relapse. After reports that adoptive immunotherapy with T-regulatory cells controls the alloreactivity of conventional T lymphocytes in animal models, tomorrow’s world of haploidentical transplantation will focus on new “designed” grafts. They will contain an appropriate ratio of conventional T lymphocytes and T-regulatory cells, natural killer cells, γ δ T cells, and other accessory cells. Preliminary results of ongoing clinical trials show the approach is feasible. It is associated with better immune reconstitution and a quite powerful graft-versus-leukemia effect with a low incidence of graft-versus-host disease and no need for posttransplant pharmacological prophylaxis. Future strategies will focus on enhancing the clinical benefit of T-regulatory cells by increasing their number and strengthening their function.
Collapse
|
260
|
Bacchetta R, Lucarelli B, Sartirana C, Gregori S, Lupo Stanghellini MT, Miqueu P, Tomiuk S, Hernandez-Fuentes M, Gianolini ME, Greco R, Bernardi M, Zappone E, Rossini S, Janssen U, Ambrosi A, Salomoni M, Peccatori J, Ciceri F, Roncarolo MG. Immunological Outcome in Haploidentical-HSC Transplanted Patients Treated with IL-10-Anergized Donor T Cells. Front Immunol 2014; 5:16. [PMID: 24550909 PMCID: PMC3907718 DOI: 10.3389/fimmu.2014.00016] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 01/09/2014] [Indexed: 11/21/2022] Open
Abstract
T-cell therapy after hematopoietic stem cell transplantation (HSCT) has been used alone or in combination with immunosuppression to cure hematologic malignancies and to prevent disease recurrence. Here, we describe the outcome of patients with high-risk/advanced stage hematologic malignancies, who received T-cell depleted (TCD) haploidentical-HSCT (haplo-HSCT) combined with donor T lymphocytes pretreated with IL-10 (ALT-TEN trial). IL-10-anergized donor T cells (IL-10-DLI) contained T regulatory type 1 (Tr1) cells specific for the host alloantigens, limiting donor-vs.-host-reactivity, and memory T cells able to respond to pathogens. IL-10-DLI were infused in 12 patients with the goal of improving immune reconstitution after haplo-HSCT without increasing the risk of graft-versus-host-disease (GvHD). IL-10-DLI led to fast immune reconstitution in five patients. In four out of the five patients, total T-cell counts, TCR-Vβ repertoire and T-cell functions progressively normalized after IL-10-DLI. These four patients are alive, in complete disease remission and immunosuppression-free at 7.2 years (median follow-up) after haplo-HSCT. Transient GvHD was observed in the immune reconstituted (IR) patients, despite persistent host-specific hypo-responsiveness of donor T cells in vitro and enrichment of cells with Tr1-specific biomarkers in vivo. Gene-expression profiles of IR patients showed a common signature of tolerance. This study provides the first indication of the feasibility of Tr1 cell-based therapy and paves way for the use of these Tr1 cells as adjuvant treatment for malignancies and immune-mediated disorders.
Collapse
Affiliation(s)
- Rosa Bacchetta
- Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, San Raffaele Telethon Institute for Gene Therapy , Milan , Italy
| | - Barbarella Lucarelli
- Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, San Raffaele Telethon Institute for Gene Therapy , Milan , Italy
| | - Claudia Sartirana
- Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, San Raffaele Telethon Institute for Gene Therapy , Milan , Italy
| | - Silvia Gregori
- Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, San Raffaele Telethon Institute for Gene Therapy , Milan , Italy
| | | | | | | | | | - Monica E Gianolini
- Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, San Raffaele Telethon Institute for Gene Therapy , Milan , Italy
| | - Raffaella Greco
- Hematology and BMT Unit, San Raffaele Hospital , Milan , Italy
| | | | | | - Silvano Rossini
- Unit of Immunohaematology and Transfusion Medicine Service, San Raffaele Hospital , Milan , Italy
| | - Uwe Janssen
- Miltenyi Biotec GmbH , Bergisch-Gladbach , Germany
| | - Alessandro Ambrosi
- Center for Statistics in Biomedical Sciences, San Raffaele Scientific Institute , Milan , Italy
| | | | | | - Fabio Ciceri
- Hematology and BMT Unit, San Raffaele Hospital , Milan , Italy
| | - Maria-Grazia Roncarolo
- Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, San Raffaele Telethon Institute for Gene Therapy , Milan , Italy ; Vita-Salute San Raffaele University , Milan , Italy
| |
Collapse
|
261
|
Kaipe H, Erkers T, Sadeghi B, Ringdén O. Stromal cells–are they really useful for GVHD? Bone Marrow Transplant 2014; 49:737-43. [DOI: 10.1038/bmt.2013.237] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 12/19/2013] [Indexed: 12/29/2022]
|
262
|
An efficient large-scale retroviral transduction method involving preloading the vector into a RetroNectin-coated bag with low-temperature shaking. PLoS One 2014; 9:e86275. [PMID: 24454964 PMCID: PMC3893289 DOI: 10.1371/journal.pone.0086275] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 12/10/2013] [Indexed: 01/15/2023] Open
Abstract
In retroviral vector-mediated gene transfer, transduction efficiency can be hampered by inhibitory molecules derived from the culture fluid of virus producer cell lines. To remove these inhibitory molecules to enable better gene transduction, we had previously developed a transduction method using a fibronectin fragment-coated vessel (i.e., the RetroNectin-bound virus transduction method). In the present study, we developed a method that combined RetroNectin-bound virus transduction with low-temperature shaking and applied this method in manufacturing autologous retroviral-engineered T cells for adoptive transfer gene therapy in a large-scale closed system. Retroviral vector was preloaded into a RetroNectin-coated bag and incubated at 4°C for 16 h on a reciprocating shaker at 50 rounds per minute. After the supernatant was removed, activated T cells were added to the bag. The bag transduction method has the advantage of increasing transduction efficiency, as simply flipping over the bag during gene transduction facilitates more efficient utilization of the retroviral vector adsorbed on the top and bottom surfaces of the bag. Finally, we performed validation runs of endoribonuclease MazF-modified CD4(+) T cell manufacturing for HIV-1 gene therapy and T cell receptor-modified T cell manufacturing for MAGE-A4 antigen-expressing cancer gene therapy and achieved over 200-fold (≥ 10(10)) and 100-fold (≥ 5 × 10(9)) expansion, respectively. In conclusion, we demonstrated that the large-scale closed transduction system is highly efficient for retroviral vector-based T cell manufacturing for adoptive transfer gene therapy, and this technology is expected to be amenable to automation and improve current clinical gene therapy protocols.
Collapse
|
263
|
Larson S, De Oliveira SN. Gene-modified hematopoietic stem cells for cancer immunotherapy. Hum Vaccin Immunother 2014; 10:982-5. [PMID: 24398603 DOI: 10.4161/hv.27637] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The rapid expansion of available cancer immunotherapies has resulted in favorable early outcomes. Specifically the use of gene therapy to introduce chimeric antigen receptors (CARs) and T cell receptors (TCRs) in T cells creates new immunotherapy options for patients. While showing early success with these approaches, limitations remain that can be overcome by the use of modification of hematopoietic stem cells (HSCs) to express CARs and TCRs. With modern gene therapy technologies, increased safety and control of the modification of the HSCs can be achieved through the use of a suicide gene.
Collapse
Affiliation(s)
- Sarah Larson
- Department of Medicine; Division of Hematology/Oncology; David Geffen School of Medicine at the University of California, Los Angeles; Los Angeles, CA USA
| | - Satiro N De Oliveira
- Department of Pediatrics; Division of Hematology/Oncology; David Geffen School of Medicine at the University of California, Los Angeles; Los Angeles, CA USA
| |
Collapse
|
264
|
Newell LF, Deans RJ, Maziarz RT. Adult adherent stromal cells in the management of graft-versus-host disease. Expert Opin Biol Ther 2014; 14:231-46. [DOI: 10.1517/14712598.2014.866648] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
265
|
DeRenzo C, Gottschalk S. Genetically modified T-cell therapy for osteosarcoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 804:323-40. [PMID: 24924183 DOI: 10.1007/978-3-319-04843-7_18] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
T-cell immunotherapy may offer an approach to improve outcomes for patients with osteosarcoma, who fail current therapies. In addition, it has the potential to reduce treatment-related complications for all patients. Generating tumor-specific T cells with conventional antigen presenting cells ex vivo is time consuming and often results in T-cell products with a low frequency of tumor-specific T cells. In addition, the generated T cells remain sensitive to the immunosuppressive tumor microenvironment. Genetic modification of T cells is one strategy to overcome these limitations. For example, T cells can be genetically modified to render them antigen specific, resistant to inhibitory factors, or increase their ability to home to tumor sites. Most genetic modification strategies have only been evaluated in preclinical models, however early phase clinical trials are in progress. In this chapter we review the current status of gene-modified T-cell therapy with special focus on osteosarcoma, highlighting potential antigenic targets, preclinical and clinical studies, and strategies to improve current T-cell therapy approaches.
Collapse
Affiliation(s)
- Christopher DeRenzo
- Center for Cell and Gene Therapy, Houston Methodist, Texas Children's Hospital, Baylor College of Medicine, 1102 Bates Street, Suite 1770, Houston, TX, 77030, USA
| | | |
Collapse
|
266
|
Yang K, Si Y, Chen H, He X, Lou J, Bai X, Du J, Guo Z, Chen P. Comparative study of the efficacy of allogeneic hematopoietic stem cell transplantation from human leukocyte antigen-haploidentical related and unrelated donors in the treatment of leukemia. Acta Haematol 2014; 131:37-44. [PMID: 24029046 DOI: 10.1159/000353776] [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: 12/13/2012] [Accepted: 06/17/2013] [Indexed: 11/19/2022]
Abstract
AIMS To compare the efficacy of hematopoietic stem cell transplantation (HSCT) from human leukocyte antigen (HLA)-haploidentical related donors (RD) and unrelated donors (URD) in the treatment of leukemia. METHODS Ninety-three leukemia patients underwent allogeneic HSCT were divided into two groups: 51 cases of RD-HSCT and 42 cases of URD-HSCT. In the RD-HSCT group, a preconditioning regimen with fludarabine (Flu) + busulfan (Bu) + cytosine arabinoside (Ara-C) was used in 42 patients and total body irradiation (TBI) + Flu + Ara-C was used in the remaining 9. RESULTS In the URD-HSCT group, a modified preconditioning regimen with Bu + cyclophosphamide was used in 35 patients, while the other 7 patients underwent treatment with TBI + Flu. After transplantation, the occurrence rate of grade II-IV acute graft-versus-host disease (GVHD) was 46.0 and 51.2% in the two groups. Likewise, the rate of chronic GVHD was 46.0 and 63.4%, respectively. No significant differences in the occurrence of acute and chronic GVHD were detected between the two groups. The differences in early-stage infection rate after transplantation, recurrence rate, 3-year survival rate, and disease-free survival rate between the two groups were not significant. CONCLUSION HLA-haploidentical RD-HSCT with enhanced preconditioning and administration of immunosuppressants showed a clinical efficacy similar to that of URD-HSCT against leukemia, without the risk of increased infection and GVHD.
Collapse
Affiliation(s)
- Kai Yang
- Department of Hematology, Beijing Military General Hospital, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
267
|
Abstract
Treatment of acute myeloid leukemia (AML) with current chemotherapy regimens is still disappointing, with overall survival rates of ≤40% at 5 years. It is now well established that AML cells can evade the immune system through multiple mechanisms, including the expression of the enzyme indoleamine 2,3 dioxygenase. Immunotherapeutic strategies, including both active, such as vaccination with leukemia-associated antigens, and passive, such as adoptive transfer of allogeneic natural killer cells, may overcome leukemia escape and lead to improved cure. Allogeneic hemopoeitic stem cell transplantation, the most effective treatment of AML, is the best known model of immunotherapy. Following transplant, recipient AML cells are eradicated by donor immune cells through the graft-versus-leukemia (GVL) effect. However, GVL is clinically associated with graft-versus-host disease, the major cause of mortality after transplant. GVL is mediated by donor T cells recognizing either leukemia-associated antigens or minor as well as major histocompatibility antigens. Several innovative strategies have been devised to generate leukemia reactive T cells so as to increase GVL responses with no or little graft-versus-host disease.
Collapse
Affiliation(s)
- Mario Arpinati
- Department of Hematology & Oncological Sciences ‘Seragnoli’, University of Bologna, Italy
| | - Antonio Curti
- Department of Hematology & Oncological Sciences ‘Seragnoli’, University of Bologna, Italy
| |
Collapse
|
268
|
Krebs S, Rodríguez-Cruz TG, Derenzo C, Gottschalk S. Genetically modified T cells to target glioblastoma. Front Oncol 2013; 3:322. [PMID: 24427741 PMCID: PMC3876295 DOI: 10.3389/fonc.2013.00322] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 12/17/2013] [Indexed: 01/21/2023] Open
Abstract
Despite advances in surgical procedures, radiation, and chemotherapy the outcome for patients with glioblastoma (GBM) remains poor. While GBM cells express antigens that are potentially recognized by T cells, GBMs prevent the induction of GBM-specific immune responses by creating an immunosuppressive microenvironment. The advent of gene transfer has allowed the rapid generation of antigen-specific T cells as well as T cells with enhanced effector function. Here we review recent advances in the field of cell therapy with genetically modified T cells and how these advances might improve outcomes for patients with GBM in the future.
Collapse
Affiliation(s)
- Simone Krebs
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine , Houston, TX , USA ; Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine , Houston, TX , USA ; Department of Pediatrics, Baylor College of Medicine , Houston, TX , USA
| | - Tania G Rodríguez-Cruz
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine , Houston, TX , USA ; Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine , Houston, TX , USA ; Department of Pediatrics, Baylor College of Medicine , Houston, TX , USA
| | - Christopher Derenzo
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine , Houston, TX , USA ; Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine , Houston, TX , USA ; Department of Pediatrics, Baylor College of Medicine , Houston, TX , USA
| | - Stephen Gottschalk
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine , Houston, TX , USA ; Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine , Houston, TX , USA ; Department of Pediatrics, Baylor College of Medicine , Houston, TX , USA ; Department of Pathology and Immunology, Baylor College of Medicine , Houston, TX , USA
| |
Collapse
|
269
|
Cheuk DKL. Optimal stem cell source for allogeneic stem cell transplantation for hematological malignancies. World J Transplant 2013; 3:99-112. [PMID: 24392314 PMCID: PMC3879529 DOI: 10.5500/wjt.v3.i4.99] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 11/15/2013] [Accepted: 12/11/2013] [Indexed: 02/05/2023] Open
Abstract
Hematopoietic stem cell transplant (HSCT) is a standard treatment for many hematological malignancies. Three different sources of stem cells, namely bone marrow (BM), peripheral blood stem cells (PBSC) and cord blood (CB) can be used for HSCT, and each has its own advantages and disadvantages. Randomized controlled trials (RCTs) suggest that there is no significant survival advantage of PBSC over BM in Human Leukocyte Antigen-matched sibling transplant for adult patients with hematological malignancies. PBSC transplant probably results in lower risk of relapse and hence better disease-free survival, especially in patients with high risk disease at the expense of higher risks of both severe acute and chronic graft-versus-host disease (GVHD). In the unrelated donor setting, the only RCT available suggests that PBSC and BM result in comparable overall and disease-free survivals in patients with hematological malignancies; and PBSC transplant results in lower risk of graft failure and higher risk of chronic GVHD. High level evidence is not available for CB in comparison to BM or PBSC. The risks and benefits of different sources of stem cells likely change with different conditioning regimen, strategies for prophylaxis and treatment of GVHD and manipulation of grafts. The recent success and rapid advance of double CB transplant and haploidentical BM and PBSC transplants further complicate the selection of stem cell source. Optimal selection requires careful weighing of the risks and benefits of different stem cell source for each individual recipient and donor. Detailed counseling of patient and donor regarding risks and benefits in the specific context of the patient and transplant method is essential for informed decision making.
Collapse
|
270
|
Adoptive T-cell therapy for hematological malignancies using T cells gene-modified to express tumor antigen-specific receptors. Int J Hematol 2013; 99:123-31. [DOI: 10.1007/s12185-013-1493-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 12/04/2013] [Accepted: 12/04/2013] [Indexed: 12/24/2022]
|
271
|
Davila ML, Bouhassira DCG, Park JH, Curran KJ, Smith EL, Pegram HJ, Brentjens R. Chimeric antigen receptors for the adoptive T cell therapy of hematologic malignancies. Int J Hematol 2013; 99:361-71. [PMID: 24311149 DOI: 10.1007/s12185-013-1479-5] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Accepted: 11/14/2013] [Indexed: 12/20/2022]
Abstract
The genetic modification of autologous T cells with chimeric antigen receptors (CARs) represents a breakthrough for gene engineering as a cancer therapy for hematologic malignancies. By targeting the CD19 antigen, we have demonstrated robust and rapid anti-leukemia activity in patients with heavily pre-treated and chemotherapy-refractory B cell acute lymphoblastic leukemia (B-ALL). We demonstrated rapid induction of deep molecular remissions in adults, which has been recently confirmed in a case report involving a child with B-ALL. In contrast to the results when treating B-ALL, outcomes have been more modest in patients with chronic lymphocytic leukemia (CLL) or other non-hodgkin's lymphoma (NHL). We review the clinical trial experience targeting B-ALL and CLL and speculate on the possible reasons for the different outcomes and propose potential optimization to CAR T cell therapy when targeting CLL or other indolent NHL. Lastly, we discuss the pre-clinical development and potential for clinical translation for using CAR T cells against multiple myeloma and acute myeloid leukemia. We highlight the potential risks and benefits by targeting these poor outcome hematologic malignancies.
Collapse
Affiliation(s)
- Marco L Davila
- Leukemia Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA,
| | | | | | | | | | | | | |
Collapse
|
272
|
Immune reconstitution after haploidentical hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 2013; 20:440-9. [PMID: 24315844 DOI: 10.1016/j.bbmt.2013.11.028] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 11/29/2013] [Indexed: 12/12/2022]
Abstract
Haploidentical hematopoietic stem cell transplantation (HSCT) offers the benefits of rapid and nearly universal donor availability and has been accepted worldwide as an alternative treatment for patients with hematologic malignancies who do not have a completely HLA-matched sibling or who require urgent transplantation. Unfortunately, serious infections and leukemia relapse resulting from slow immune reconstitution remain the 2 most frequent causes of mortality in patients undergoing haploidentical HSCT, particularly in those receiving extensively T cell-depleted megadose CD34(+) allografts. This review summarizes advances in immune recovery after haploidentical HSCT, focusing on the immune subsets likely to have the greatest impact on clinical outcomes. The progress made in accelerating immune reconstitution using different strategies after haploidentical HSCT is also discussed. It is our belief that a predictive immune subset-guided strategy to improve immune recovery might represent a future clinical direction.
Collapse
|
273
|
Engineered T cells for cancer treatment. Cytotherapy 2013; 16:713-33. [PMID: 24239105 DOI: 10.1016/j.jcyt.2013.10.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 10/01/2013] [Accepted: 10/05/2013] [Indexed: 01/08/2023]
Abstract
Adoptively transferred T cells have the capacity to traffic to distant tumor sites, infiltrate fibrotic tissue and kill antigen-expressing tumor cells. Various groups have investigated different genetic engineering strategies designed to enhance tumor specificity, increase T cell potency, improve proliferation, persistence or migratory capacity and increase safety. This review focuses on recent developments in T cell engineering, discusses the clinical application of these engineered cell products and outlines future prospects for this therapeutic modality.
Collapse
|
274
|
Locatelli F, Lucarelli B, Merli P. Current and future approaches to treat graft failure after allogeneic hematopoietic stem cell transplantation. Expert Opin Pharmacother 2013; 15:23-36. [DOI: 10.1517/14656566.2014.852537] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
275
|
Zhan H, Gilmour K, Chan L, Farzaneh F, McNicol AM, Xu JH, Adams S, Fehse B, Veys P, Thrasher A, Gaspar H, Qasim W. Production and first-in-man use of T cells engineered to express a HSVTK-CD34 sort-suicide gene. PLoS One 2013; 8:e77106. [PMID: 24204746 PMCID: PMC3804528 DOI: 10.1371/journal.pone.0077106] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 08/29/2013] [Indexed: 11/22/2022] Open
Abstract
Suicide gene modified donor T cells can improve immune reconstitution after allogeneic haematopoietic stem cell transplantation (SCT), but can be eliminated in the event of graft versus host disease (GVHD) through the administration of prodrug. Here we report the production and first-in-man use of mismatched donor T cells modified with a gamma-retroviral vector expressing a herpes simplex thymidine kinase (HSVTK):truncated CD34 (tCD34) suicide gene/magnetic selection marker protein. A stable packaging cell line was established to produce clinical grade vector pseudotyped with the Gibbon Ape Leukaemia Virus (GALV). T cells were transduced in a closed bag system following activation with anti-CD3/CD28 beads, and enriched on the basis of CD34 expression. Engineered cells were administered in two escalating doses to three children receiving T-depleted, CD34 stem cell selected, mismatched allogeneic grafts. All patients had pre-existing viral infections and received chemotherapy conditioning without serotherapy. In all three subjects cell therapy was tolerated without acute toxicity or the development of acute GVHD. Circulating gene modified T cells were detectable by flow cytometry and by molecular tracking in all three subjects. There was resolution of virus infections, concordant with detectable antigen-specific T cell responses and gene modified cells persisted for over 12 months. These findings highlight the suitability of tCD34 as a GMP compliant selection marker and demonstrate the feasibility, safety and immunological potential of HSVTK-tCD34 suicide gene modified donor T cells. Trial Registration ClinicalTrials.gov NCT01204502 <NCT01204502>
Collapse
Affiliation(s)
- Hong Zhan
- Molecular Immunology Unit, Institute of Child Health (ICH), University College London (UCL), London, United Kingdom
| | - Kimberly Gilmour
- Molecular Immunology Unit, Institute of Child Health (ICH), University College London (UCL), London, United Kingdom
| | - Lucas Chan
- Department of Haematological Medicine, The Rayne institute, Kings College London (KCL), London, United Kingdom
| | - Farzin Farzaneh
- Department of Haematological Medicine, The Rayne institute, Kings College London (KCL), London, United Kingdom
| | - Anne Marie McNicol
- Molecular Immunology Unit, Institute of Child Health (ICH), University College London (UCL), London, United Kingdom
| | - Jin-Hua Xu
- Molecular Immunology Unit, Institute of Child Health (ICH), University College London (UCL), London, United Kingdom
| | - Stuart Adams
- Molecular Immunology Unit, Institute of Child Health (ICH), University College London (UCL), London, United Kingdom
| | - Boris Fehse
- Bone Marrow Transplant Unit, University of Hamburg, Hamburg, Germany
| | - Paul Veys
- Molecular Immunology Unit, Institute of Child Health (ICH), University College London (UCL), London, United Kingdom
| | - Adrian Thrasher
- Molecular Immunology Unit, Institute of Child Health (ICH), University College London (UCL), London, United Kingdom
| | - Hubert Gaspar
- Molecular Immunology Unit, Institute of Child Health (ICH), University College London (UCL), London, United Kingdom
| | - Waseem Qasim
- Molecular Immunology Unit, Institute of Child Health (ICH), University College London (UCL), London, United Kingdom
- * E-mail:
| |
Collapse
|
276
|
Abstract
Gene transfer vectors derived from oncoretroviruses or lentiviruses are the most robust and reliable tools to stably integrate therapeutic transgenes in human cells for clinical applications. Integration of these vectors in the genome may, however, have undesired effects caused by insertional deregulation of gene expression at the transcriptional or post-transcriptional level. The occurrence of severe adverse events in several clinical trials involving the transplantation of stem cells genetically corrected with retroviral vectors showed that insertional mutagenesis is not just a theoretical event, and that retroviral transgenesis is associated with a finite risk of genotoxicity. In addressing these issues, the gene therapy community offered a spectacular example of how scientific knowledge and technology can be put to work to understand the causes of unpredicted side effects, design new vectors, and develop tools and models to predict their safety and efficacy. As an added benefit, these efforts brought new basic knowledge on virus-host interactions and on the biology and dynamics of human somatic stem cells. This review summarizes the current knowledge on the interactions between retroviruses and the human genome and addresses the impact of target site selection on the safety of retroviral vector-mediated gene therapy.
Collapse
Affiliation(s)
- Alessia Cavazza
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | | | | |
Collapse
|
277
|
CD44v6-targeted T cells mediate potent antitumor effects against acute myeloid leukemia and multiple myeloma. Blood 2013; 122:3461-72. [PMID: 24016461 DOI: 10.1182/blood-2013-04-493361] [Citation(s) in RCA: 282] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Genetically targeted T cells promise to solve the feasibility and efficacy hurdles of adoptive T-cell therapy for cancer. Selecting a target expressed in multiple-tumor types and that is required for tumor growth would widen disease indications and prevent immune escape caused by the emergence of antigen-loss variants. The adhesive receptor CD44 is broadly expressed in hematologic and epithelial tumors, where it contributes to the cancer stem/initiating phenotype. In this study, silencing of its isoform variant 6 (CD44v6) prevented engraftment of human acute myeloid leukemia (AML) and multiple myeloma (MM) cells in immunocompromised mice. Accordingly, T cells targeted to CD44v6 by means of a chimeric antigen receptor containing a CD28 signaling domain mediated potent antitumor effects against primary AML and MM while sparing normal hematopoietic stem cells and CD44v6-expressing keratinocytes. Importantly, in vitro activation with CD3/CD28 beads and interleukin (IL)-7/IL-15 was required for antitumor efficacy in vivo. Finally, coexpressing a suicide gene enabled fast and efficient pharmacologic ablation of CD44v6-targeted T cells and complete rescue from hyperacute xenogeneic graft-versus-host disease modeling early and generalized toxicity. These results warrant the clinical investigation of suicidal CD44v6-targeted T cells in AML and MM.
Collapse
|
278
|
Abstract
Immune suppression is a risk factor for malignant lymphoma development. Progress in medical science has increased the numbers of immunosuppressed patients due to organ transplantations or successful treatment of autoimmune diseases. Different forms of immune suppression and the respective lymphoma entities are discussed in this article. Another issue treated are gray zone lymphomas between Hodgkin's lymphoma and diffuse large B cell lymphoma. This category not only represents a diagnostic challenge but also represents more a true biological continuum.
Collapse
Affiliation(s)
- S Hartmann
- Senckenbergisches Institut für Pathologie, Klinikum der J. W. Goethe-Universität, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Deutschland.
| | | |
Collapse
|
279
|
Linn YC. Adoptive immunotherapy with polyclonal T cells and natural killer cells for hematological malignancies: current status and future prospects. Int J Hematol Oncol 2013. [DOI: 10.2217/ijh.13.31] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
SUMMARY Adoptive cellular therapy with polyclonal T cells and natural killer cells are immunotherapeutic modalities being studied in solid tumors and hematological malignancies to treat disease and prevent relapse. These include unexpanded polyclonal T cells, short-term activation by cytokine into lymphokine-activated killer cells, longer term expansion by cytokine stimulation giving rise to cytokine-induced killer cells or expansion under costimulation with beads expressing anti-CD3 and anti-CD28. Similarly natural killer cells can be given with or without activation and expansion. Here we review the published work and clinical trials involving each cell type in the autologous, matched allogeneic, haploidentical and nontransplant settings, comparing and contrasting each cell type and discussing their potential applications.
Collapse
Affiliation(s)
- Yeh-Ching Linn
- Department of Hematology, Singapore General Hospital, Outram Road, Singapore 169608
| |
Collapse
|
280
|
Qian L, Wu Z, Shen J. Advances in the treatment of acute graft-versus-host disease. J Cell Mol Med 2013; 17:966-975. [PMID: 23802653 PMCID: PMC3780546 DOI: 10.1111/jcmm.12093] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 05/15/2013] [Indexed: 12/13/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) has been widely used for the treatment of hematologic malignant and non-malignant hematologic diseases and other diseases. However, acute graft-versus-host disease (GVHD) is a life-threatening complication of allogeneic transplantation. Acute GVHD may occur in 30% of transplant recipients, which is a syndrome of erythematous skin eruption, cholestatic liver disease and intestinal dysfunction, resulting from the activation of donor T lymphocytes by host antigen-presenting cells, resulting in an immune-mediated inflammatory response. Recent scientific advances in the understanding of the pathogenesis involved in the development of acute GVHD and clinical investigation have provided more effective therapeutic strategies for acute GVHD. This review focuses on major scientific and clinical advances in the treatment of acute GVHD.
Collapse
Affiliation(s)
- Liren Qian
- Department of Haematology, Navy General HospitalBeijing, China
| | - Zhengcheng Wu
- Department of Neurology, Navy General HospitalBeijing, China
| | - Jianliang Shen
- Department of Haematology, Navy General HospitalBeijing, China
| |
Collapse
|
281
|
The CIBMTR score predicts survival of AML patients undergoing allogeneic transplantation with active disease after a myeloablative or reduced intensity conditioning: a retrospective analysis of the Gruppo Italiano Trapianto Di Midollo Osseo. Leukemia 2013; 27:2086-91. [PMID: 23835862 PMCID: PMC3806246 DOI: 10.1038/leu.2013.208] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
282
|
Haploidentical transplantation for hematologic malignancies: where do we stand? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2013; 2012:230-6. [PMID: 23233586 DOI: 10.1182/asheducation-2012.1.230] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The fundamental obstacle to the successful application of partially HLA-mismatched related donor, or HLA-haploidentical stem cell transplantation, is the strength of the host and donor T-cell response to allogeneic HLA molecules, which results in increased incidences of graft failure, GVHD, and nonrelapse mortality. The holy grail of haplo-SCT is to mitigate host-versus-graft and graft-versus-host responses while preserving immune responses to infection and the patient's malignancy. Two strategies have been taken to achieve this goal. The first strategy is to supplement a T cell-depleted graft with pathogen-specific T cells or populations of T cells in which alloreactivity can be controlled. The second strategy is to eliminate alloreactive T cells selectively from a T cell-replete graft. Substantial progress has been made with both approaches so that the safety of haplo-SCT now approaches that of SCT using grafts of umbilical cord blood or from HLA-matched donors. In light of the rapid and near universal availability of HLA-haploidentical related donors, it should now be possible to identify and mobilize a donor for every patient referred for allogeneic SCT. Prospective comparisons between haploidentical SCT and unrelated donor SCT should be performed to identify the most efficacious approach to alternative donor transplantation.
Collapse
|
283
|
Booth C, Veys P. T cell depletion in paediatric stem cell transplantation. Clin Exp Immunol 2013; 172:139-47. [PMID: 23574311 DOI: 10.1111/cei.12004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2012] [Indexed: 01/25/2023] Open
Abstract
Haematopoietic stem cell transplantation (HSCT) can be a curative procedure for a growing number of paediatric diseases, but as the indications for HSCT grow, so does the need to find suitable stem cell donors. When the preferred option of a genoidentical sibling donor is not available alternative donors, including unrelated adult or umbilical cord blood donors, or haploidentical related donors may be considered. Outcome following alternative donor HSCT has improved over the past 20 years but graft-versus-host disease (GvHD) remains a significant obstacle. T cell depletion (TCD) for non-genoidentical grafts aims to reduce the morbidity and mortality associated with GvHD, but this intervention has not led directly to improved survival due to delayed immune reconstitution and increased infections, graft rejection and increased rates of disease relapse. Limited data from the paediatric population, however, suggest some encouraging results for children undergoing haploidentical HSCT: a move from positive selection of CD34(+) haematopoietic stem cells towards negative depletion of specific cell subsets in order to retain useful accessory cells within the graft appears to enhance immune reconstitution and improve disease-free survival. Here we review recent paediatric outcome data for T cell-depleted HSCT, explore the role of serotherapy in conditioning regimens and look at future possibilities to improve outcome, including novel allodepletion techniques, suicide gene therapy and pathogen-specific immunotherapy.
Collapse
Affiliation(s)
- C Booth
- Institute of Child Health, University College London, London, UK.
| | | |
Collapse
|
284
|
Arber C, Brenner MK, Reddy P. Mouse models in bone marrow transplantation and adoptive cellular therapy. Semin Hematol 2013; 50:131-44. [PMID: 24216170 DOI: 10.1053/j.seminhematol.2013.03.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Mouse models of transplantation have been indispensable to the development of bone marrow transplantation (BMT). Their role in the generation of basic science knowledge is invaluable and is subject to discussion below. However, this article focuses on the direct role and relevance of mouse models towards the clinical development and advances in BMT and adoptive T-cell therapy for human diseases. The authors aim to present a thoughtful perspective on the pros and cons of mouse models while noting that despite imperfections these models are obligatory for the development of science-based medicine.
Collapse
Affiliation(s)
- Caroline Arber
- Center for Cell and Gene Therapy, Baylor College of Medicine, The Methodist Hospital and Texas Children's Hospital, Houston, TX
| | | | | |
Collapse
|
285
|
T lymphocytes are not immune. Mol Ther 2013; 21:1114-5. [PMID: 23728254 DOI: 10.1038/mt.2013.102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
286
|
Aiuti A, Cossu G, de Felipe P, Galli MC, Narayanan G, Renner M, Stahlbom A, Schneider CK, Voltz-Girolt C. The Committee for Advanced Therapies' of the European Medicines Agency Reflection Paper on Management of Clinical Risks Deriving from Insertional Mutagenesis. HUM GENE THER CL DEV 2013; 24:47-54. [DOI: 10.1089/humc.2013.119] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Alessandro Aiuti
- Gene Therapy Working Party of the Committee for Advanced Therapies, European Medicines Agency, 7, Westferry Circus E14 4HB, London, United Kingdom
- San Raffaele-Telethon Institute for Gene Therapy, Via Olgettina 58, 20132 Milan, Italy
- University Department of Pediatrics, Bambino Gesù Children's Hospital and Tor Vergata University, Rome, Italy
| | - Giulio Cossu
- Committee for Advanced Therapies, European Medicines Agency, 7, Westferry Circus E14 4HB, London, United Kingdom
- Department of Cell and Development Biology, University College London, Gower Street, London, WC1E 6BT United Kingdom
| | - Pablo de Felipe
- Gene Therapy Working Party of the Committee for Advanced Therapies, European Medicines Agency, 7, Westferry Circus E14 4HB, London, United Kingdom
- Agencia Española de Medicamentos y Productos Sanitarios, Madrid, Spain
| | - Maria Cristina Galli
- Gene Therapy Working Party of the Committee for Advanced Therapies, European Medicines Agency, 7, Westferry Circus E14 4HB, London, United Kingdom
- Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy
| | - Gopalan Narayanan
- Committee for Advanced Therapies, European Medicines Agency, 7, Westferry Circus E14 4HB, London, United Kingdom
- Medicines and Healthcare Products Regulatory Agency, 151 Buckingham Palace Road, Victoria, London, SW1W 9SZ United Kingdom
| | - Matthias Renner
- Gene Therapy Working Party of the Committee for Advanced Therapies, European Medicines Agency, 7, Westferry Circus E14 4HB, London, United Kingdom
- Paul-Ehrlich-Institut, Paul-Ehrlich-Str. 51-59, D-63225 Langen, Germany
| | - Axel Stahlbom
- Gene Therapy Working Party of the Committee for Advanced Therapies, European Medicines Agency, 7, Westferry Circus E14 4HB, London, United Kingdom
- Läkemedelsverket (Medical Products Agency), Dag Hammarskjölds väg 42, 75103 Uppsala, Sweden
| | - Christian K. Schneider
- Committee for Advanced Therapies, European Medicines Agency, 7, Westferry Circus E14 4HB, London, United Kingdom
- Danish Health and Medicines Authority, Axel Heides Gade 1, 2300 Copenhagen, Denmark
- Twincore Centre for Experimental and Clinical Infection Research, Feodor-Lynen-Straße 730625 Hannover, Germany
| | - Caroline Voltz-Girolt
- Gene Therapy Working Party of the Committee for Advanced Therapies, European Medicines Agency, 7, Westferry Circus E14 4HB, London, United Kingdom
| |
Collapse
|
287
|
Booth C, Lawson S, Veys P. The current role of T cell depletion in paediatric stem cell transplantation. Br J Haematol 2013; 162:177-90. [DOI: 10.1111/bjh.12400] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 03/07/2013] [Indexed: 02/03/2023]
Affiliation(s)
- Claire Booth
- Molecular Immunology Unit; Institute of Child Health; University College London; London UK
| | - Sarah Lawson
- Department of Haematology; Birmingham Children's Hospital NHS Foundation Trust; Birmingham UK
| | - Paul Veys
- Molecular Immunology Unit; Institute of Child Health; University College London; London UK
- Department of Blood and Marrow Transplantation; Great Ormond Street Hospital for Children NHS Foundation Trust; London UK
| |
Collapse
|
288
|
The immunogenicity of virus-derived 2A sequences in immunocompetent individuals. Gene Ther 2013; 20:958-62. [PMID: 23698740 DOI: 10.1038/gt.2013.25] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Revised: 03/21/2013] [Accepted: 04/17/2013] [Indexed: 12/31/2022]
Abstract
Genetic engineering of T cells for adoptive immunotherapy in cancer patients has shown significant promise. To ensure optimal antitumor activity and safety, the simultaneous expression of multiple genes is frequently required, and short viral-derived 2A sequences are increasingly preferred for this purpose. Concerns exist, however, that these virus-derived sequences may induce unwanted immune responses, and thus diminish persistence of the gene-modified cells after adoptive transfer. Whereas such responses were absent in immunocompromised recipients, potential immunogenicity in immunocompetent individuals remains a concern. We now address whether ex vivo T cell responses can be elicited against the most widely used 2A sequences (2A-Thosea asigna virus (TAV) or 2A-equine rhinitis virus (ERAV), specifically) in immunocompetent individuals. We used a potent ex vivo culture system previously validated to induce T cell responses even against weakly immunogenic antigens. Of the sixteen donors tested, only five released very low levels of interferon-γ in response to 2A-TAV peptide mixtures (single peptide specificity in three donors, adjacent self-antigen peptide specificity in one donor and nonspecific reactivity in one donor). None of them produced cytotoxic activity or responded to 2A-ERAV. These results suggest that exposure to viral-derived 2A sequences is unlikely to produce unwanted T cell responses in immunocompetent individuals and further supports their continued use for studies of human gene therapy.
Collapse
|
289
|
Abstract
PURPOSE OF REVIEW This article reviews the impact of natural killer (NK) cell alloreactivity on hematopoietic cell transplantation since it was first observed in haploidentical transplant recipients 10 years ago. RECENT FINDINGS Research has established 'missing self-recognition' as the mechanism underlying NK cell-mediated graft-versus-leukemia effects in T-cell-depleted haploidentical hematopoietic cell transplantation and has clarified optimal transplantation protocols to harness NK cell alloreactivity. SUMMARY In the past decade, clinical studies have shown that the benefits of donor-versus-recipient NK cell alloreactivity in haploidentical transplantation are triggered by specific human leukocyte antigen (HLA) class I mismatches. Donor HLA is crucial for driving NK cell education so that reconstituting NK cells mature as donor-tolerant and recipient-alloreactive. Transplantation of large doses of extensively T-cell-depleted hematopoietic grafts with no posttransplant immune suppression was found to be essential for development of NK cell alloreactivity. Clinical trials demonstrated that donor-versus-recipient NK cell alloreactivity is a key therapeutic element in haploidentical transplants for acute myeloblastic leukemia in adults and acute lymphoblastic leukemia in children. Moreover, in pilot studies, mature haploidentical NK cells were transiently transferred into lymphoablated patients with acute leukemia in remission. The results showed NK cell therapy may be a promising strategy for consolidating leukemia remission. In line with the notion that NK cell function is regulated by a balance between activating and inhibitory receptors, in the matched transplant setting, transplantation from donors possessing certain activating NK receptors (activating killer cell immunoglobulin-like receptors) appeared to protect from relapse and improved survival.
Collapse
|
290
|
Kaloyannidis P, Leen AM, Papadopoulou A. T-cell therapy: a powerful tool for the management of viral infections and relapse post hematopoietic stem cell transplantation. Expert Rev Hematol 2013; 5:471-3. [PMID: 23146049 DOI: 10.1586/ehm.12.38] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
291
|
Rager A, Porter DL. Cellular therapy following allogeneic stem-cell transplantation. Ther Adv Hematol 2013; 2:409-28. [PMID: 23556106 DOI: 10.1177/2040620711412416] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Allogeneic hematopoietic stem-cell transplantation (HSCT) is the most effective approach for many patients with hematologic malignancies. Unfortunately, relapse remains the most common cause of death after allogeneic HSCT, and the prognosis of relapsed disease is poor for most patients. Induction of a graft-versus-leukemia (GVL), or graft-versus-tumor, effect through the use of donor leukocyte infusion (DLI), or donor lymphocyte infusion, has been remarkably successful for relapsed chronic myelogenous leukemia. Unfortunately, response to DLI in other hematologic malignancies is much less common and depends on many factors including histology, pace and extent of relapse, and time from HSCT to relapse. Furthermore, graft-versus-host disease (GVHD) is common after DLI and often limits successful immunotherapy. Ultimately, manipulations to minimize GVHD while preserving or enhancing GVL are necessary to improve outcomes for relapse after allogeneic HSCT.
Collapse
|
292
|
Digiusto DL, Kiem HP. Current translational and clinical practices in hematopoietic cell and gene therapy. Cytotherapy 2013; 14:775-90. [PMID: 22799276 DOI: 10.3109/14653249.2012.694420] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Clinical trials over the last 15 years have demonstrated that cell and gene therapies for cancer, monogenic and infectious disease are feasible and can lead to long-term benefit for patients. However, these trials have been limited to proof-of-principle and were conducted on modest numbers of patients or over long periods of time. In order for these studies to move towards standard practice and commercialization, scalable technologies for the isolation, ex vivo manipulation and delivery of these cells to patients must be developed. Additionally, regulatory strategies and clinical protocols for the collection, creation and delivery of cell products must be generated. In this article we review recent progress in hematopoietic cell and gene therapy, describe some of the current issues facing the field and discuss clinical, technical and regulatory approaches used to navigate the road to product development.
Collapse
Affiliation(s)
- David L Digiusto
- Department of Virology and Laboratory for Cellular Medicine, Beckman Research Institute of the City of Hope, Duarte, California 91010, USA.
| | | |
Collapse
|
293
|
Vago L, Toffalori C, Ciceri F, Fleischhauer K. Genomic loss of mismatched human leukocyte antigen and leukemia immune escape from haploidentical graft-versus-leukemia. Semin Oncol 2013. [PMID: 23206847 DOI: 10.1053/j.seminoncol.2012.09.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Recent developments in cell processing and immunosuppressive strategies has allowed the safe infusion of high numbers of donor T cells in the context of clinical haploidentical hematopoietic stem cell transplantation (HSCT). Haploidentical T cells display an intrinsic ability to recognize and eliminate residual patient leukemic cells, largely due to alloreactivity against the patient-specific human leukocyte antigen (HLA) molecules encoded on the mismatched haplotype. However, recent evidence has shown that leukemia, like many other tumors displaying pronounced genomic instability, is frequently able to evade this potent graft-versus-leukemia effect by undergoing de novo genomic mutations, which result in the permanent loss of only those HLA molecules targeted by haploidentical donor T-cell alloreactivity. This review summarizes the recent clinical and experimental evidence regarding this phenomenon, and its therapeutic and clinical consequences.
Collapse
Affiliation(s)
- Luca Vago
- Hematology and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, Milano, Italy.
| | | | | | | |
Collapse
|
294
|
Ciceri F, Lupo-Stanghellini MT, Korthof ET. Haploidentical transplantation in patients with acquired aplastic anemia. Bone Marrow Transplant 2013; 48:183-5. [PMID: 23292235 DOI: 10.1038/bmt.2012.231] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Haploidentical SCT (haplo-SCT) has been considered a therapeutic option in patients with acquired severe aplastic anemia (SAA) failing at least one course of immune suppressive therapy with antithymocyte globulin and lacking an HLA-matched related or unrelated donor. The platforms of both ex vivo T-cell-depleted and unmanipulated grafts have been explored in children and adults. Overall, the primary objective of a stable haploidentical hematopoietic engraftment with a low rate of GVHD is unmet in a significant proportion of patients undergoing haplo-SCT for SAA. Haploidentical transplants for refractory SAA should be performed in a specialist center with major experience in hematopoietic SCT procedures and preferably performed within the framework of a local clinical protocol designed specifically to address the prevention of graft rejection and GVHD.
Collapse
Affiliation(s)
- F Ciceri
- Hematology and BMT Unit, EBMT CIC 813, San Raffaele Scientific Institute, Milano, Italy.
| | | | | |
Collapse
|
295
|
Fernandez Vina M, Heslop HE, Barker JN. New Approaches in Alternative Donor Transplantation. Biol Blood Marrow Transplant 2013; 19:S91-6. [DOI: 10.1016/j.bbmt.2012.10.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
296
|
Bire S, Rouleux-Bonnin F. Transgene Site-Specific Integration: Problems and Solutions. SITE-DIRECTED INSERTION OF TRANSGENES 2013. [DOI: 10.1007/978-94-007-4531-5_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
297
|
Park JH, Brentjens RJ. Immunotherapies in CLL. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 792:241-57. [PMID: 24014300 DOI: 10.1007/978-1-4614-8051-8_11] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Chronic lymphocytic leukemia (CLL) is the most frequently diagnosed leukemia in the Western world, yet remains essentially incurable. Although initial chemotherapy response rates are high, patients invariably relapse and subsequently develop resistance to chemotherapy. For the moment, allogeneic hematopoietic stem cell transplant (allo-HSCT) remains the only potentially curative treatment for patients with CLL, but it is associated with high rates of treatment-related mortality. Immune-based treatment strategies to augment the cytotoxic potential of T cells offer exciting new treatment options for patients with CLL, and provide a unique and powerful spectrum of tools distinct from traditional chemotherapy. Among the most novel and promising of these approaches are chimeric antigen receptor (CAR)-based cell therapies that combine advances in genetic engineering and adoptive immunotherapy.
Collapse
Affiliation(s)
- Jae H Park
- Department of Medicine, Leukemia Service, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | | |
Collapse
|
298
|
Abstract
Abstract
The fundamental obstacle to the successful application of partially HLA-mismatched related donor, or HLA-haploidentical stem cell transplantation, is the strength of the host and donor T-cell response to allogeneic HLA molecules, which results in increased incidences of graft failure, GVHD, and nonrelapse mortality. The holy grail of haplo-SCT is to mitigate host-versus-graft and graft-versus-host responses while preserving immune responses to infection and the patient's malignancy. Two strategies have been taken to achieve this goal. The first strategy is to supplement a T cell–depleted graft with pathogen-specific T cells or populations of T cells in which alloreactivity can be controlled. The second strategy is to eliminate alloreactive T cells selectively from a T cell–replete graft. Substantial progress has been made with both approaches so that the safety of haplo-SCT now approaches that of SCT using grafts of umbilical cord blood or from HLA-matched donors. In light of the rapid and near universal availability of HLA-haploidentical related donors, it should now be possible to identify and mobilize a donor for every patient referred for allogeneic SCT. Prospective comparisons between haploidentical SCT and unrelated donor SCT should be performed to identify the most efficacious approach to alternative donor transplantation.
Collapse
|
299
|
Développement des greffes haplo-identiques chez l’adulte atteint d’une hémopathie maligne. Bull Cancer 2012; 99:1141-51. [DOI: 10.1684/bdc.2012.1665] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
300
|
Aversa F, Martelli MF, Velardi A. Haploidentical Hematopoietic Stem Cell Transplantation With a Megadose T-Cell–Depleted Graft: Harnessing Natural and Adaptive Immunity. Semin Oncol 2012. [DOI: 10.1053/j.seminoncol.2012.09.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|