51
|
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
|
52
|
Wang D, Huang XF, Hong B, Song XT, Hu L, Jiang M, Zhang B, Ning H, Li Y, Xu C, Lou X, Li B, Yu Z, Hu J, Chen J, Yang F, Gao H, Ding G, Liao L, Rollins L, Jones L, Chen SY, Chen H. Efficacy of intracellular immune checkpoint-silenced DC vaccine. JCI Insight 2018; 3:98368. [PMID: 29415891 PMCID: PMC5821183 DOI: 10.1172/jci.insight.98368] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 12/28/2017] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND DC-based tumor vaccines have had limited clinical success thus far. SOCS1, a key inhibitor of inflammatory cytokine signaling, is an immune checkpoint regulator that limits DC immunopotency. METHODS We generated a genetically modified DC (gmDC) vaccine to perform immunotherapy. The adenovirus (Ad-siSSF) delivers two tumor-associated antigens (TAAs), survivin and MUC1; secretory bacterial flagellin for DC maturation; and an RNA interference moiety to suppress SOCS1. A 2-stage phase I trial was performed for patients with relapsed acute leukemia after allogenic hematopoietic stem cell transplantation: in stage 1, we compared the safety and efficacy between gmDC treatment (23 patients) and standard donor lymphocyte infusion (25 patients); in stage 2, we tested the efficacy of the gmDC vaccine for 12 acute myeloid leukemia (AML) patients with early molecular relapse. RESULTS gmDCs elicited potent TAA-specific CTL responses in vitro, and the immunostimulatory activity of gmDC vaccination was demonstrated in rhesus monkeys. A stage 1 study established that this combinatory gmDC vaccine is safe in acute leukemia patients and yielded improved survival rate. In stage 2, we observed a complete remission rate of 83% in 12 relapsed AML patients. Overall, no grade 3 or grade 4 graft-versus-host disease incidence was detected in any of the 35 patients enrolled. CONCLUSIONS This study, with combinatory modifications in DCs, demonstrates the safety and efficacy of SOCS1-silenced DCs in treating relapsed acute leukemia. TRIAL REGISTRATION ClinicalTrials.gov NCT01956630. FUNDING National Institute of Health (R01CA90427); the Key New Drug Development and Manufacturing Program of the "Twelfth Five-Year Plan" of China (2011ZX09102-001-29); and Clinical Application Research of Beijing (Z131107002213148).
Collapse
MESH Headings
- Adenoviridae/genetics
- Adolescent
- Adult
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/adverse effects
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Cell Engineering/methods
- Child
- Dendritic Cells/immunology
- Dendritic Cells/transplantation
- Female
- Follow-Up Studies
- Genetic Vectors/genetics
- Graft vs Host Disease/epidemiology
- Graft vs Host Disease/immunology
- Hematopoietic Stem Cell Transplantation/adverse effects
- Humans
- Immunotherapy, Adoptive/methods
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/therapy
- Lymphocyte Transfusion
- Male
- Middle Aged
- Neoplasm Recurrence, Local/immunology
- Neoplasm Recurrence, Local/mortality
- Neoplasm Recurrence, Local/therapy
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy
- Survival Analysis
- Transplantation, Autologous
- Treatment Outcome
- Young Adult
Collapse
Affiliation(s)
- Danhong Wang
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
| | - Xue F. Huang
- Department of Molecular Microbiology and Immunology and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| | - Bangxing Hong
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Xiao-Tong Song
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Liangding Hu
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
| | - Min Jiang
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
| | - Bin Zhang
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
| | - Hongmei Ning
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
| | - Yuhang Li
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
| | - Chen Xu
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
| | - Xiao Lou
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
| | - Botao Li
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
| | - Zhiyong Yu
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
| | - Jiangwei Hu
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
| | - Jianlin Chen
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
| | - Fan Yang
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
| | - Haiyan Gao
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
| | - Guoliang Ding
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
| | - Lianming Liao
- Department of Oncology, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Lisa Rollins
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Lindsey Jones
- Department of Molecular Microbiology and Immunology and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| | - Si-Yi Chen
- Department of Molecular Microbiology and Immunology and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Hu Chen
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|
53
|
Loeff FC, Falkenburg JHF, Hageman L, Huisman W, Veld SAJ, van Egmond HME, van de Meent M, von dem Borne PA, Veelken H, Halkes CJM, Jedema I. High Mutation Frequency of thePIGAGene in T Cells Results in Reconstitution of GPI Anchor−/CD52−T Cells That Can Give Early Immune Protection after Alemtuzumab-Based T Cell–Depleted Allogeneic Stem Cell Transplantation. THE JOURNAL OF IMMUNOLOGY 2018; 200:2199-2208. [DOI: 10.4049/jimmunol.1701018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 01/09/2018] [Indexed: 12/30/2022]
|
54
|
Karpanen T, Olweus J. The Potential of Donor T-Cell Repertoires in Neoantigen-Targeted Cancer Immunotherapy. Front Immunol 2017; 8:1718. [PMID: 29321773 PMCID: PMC5732232 DOI: 10.3389/fimmu.2017.01718] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/21/2017] [Indexed: 12/30/2022] Open
Abstract
T cells can recognize peptides encoded by mutated genes, but analysis of tumor-infiltrating lymphocytes suggests that very few neoantigens spontaneously elicit T-cell responses. This may be an important reason why immune checkpoint inhibitors are mainly effective in tumors with a high mutational burden. Reasons for clinically insufficient responses to neoantigens might be inefficient priming, inhibition, or deletion of the cognate T cells. Responses can be dramatically improved by cancer immunotherapy such as checkpoint inhibition, but often with temporary effects. By contrast, T cells from human leukocyte antigen (HLA)-matched donors can cure diseases such as chronic myeloid leukemia. The therapeutic effect is mediated by donor T cells recognizing polymorphic peptides for which the donor and patient are disparate, presented on self-HLA. Donor T-cell repertoires are unbiased by the immunosuppressive environment of the tumor. A recent study demonstrated that T cells from healthy individuals are able to respond to neoantigens that are ignored by tumor-infiltrating T cells of melanoma patients. In this review, we discuss possible reasons why neoantigens escape host T cells and how these limitations may be overcome by utilization of donor-derived T-cell repertoires to facilitate rational design of neoantigen-targeted immunotherapy.
Collapse
Affiliation(s)
- Terhi Karpanen
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, and K.G. Jebsen Center for Cancer Immunotherapy, University of Oslo, Oslo, Norway
| | - Johanna Olweus
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, and K.G. Jebsen Center for Cancer Immunotherapy, University of Oslo, Oslo, Norway
| |
Collapse
|
55
|
Mohammadi S, Norooznezhad AH, Mohammadi AM, Nasiri H, Nikbakht M, Saki N, Vaezi M, Alimoghaddam K, Ghavamzadeh A. Optimizing peripheral blood stem cells transplantation outcome through amend relapse and graft failure: a review of current literature. Exp Hematol Oncol 2017; 6:24. [PMID: 28808609 PMCID: PMC5550945 DOI: 10.1186/s40164-017-0082-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 07/24/2017] [Indexed: 11/17/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) has been considered as a valuable approach in treatment of numerous malignant and none malignant hematologic disorders. However, relapse and poor graft function (PGF) after allo-SCT remain to be controversial issues which may affect the transplantation outcome. Relevant articles were searched in MEDLINE database (2000–2016) using keywords and phrases: donor lymphocyte infusions, allogeneic stem cells transplantation, relapsed hematologic malignancies, booster schedules, cell dose, laboratory monitoring protocols and technical aspects of apheresis. Relapse of disease and PGF could be reduced via noting some main points such as choosing the suitable time and patient for donor lymphocyte infusion (DLI) and also determination of patients who ought to candidate for second allogeneic HSCT or for the use of stem cell boost. DLI and stem cell booster are promising treatment strategies noted in this review. Finally, this paper discusses indications and technical aspects of DLI and stem cell booster in hematological malignancies and emphasizes their therapeutic or pre-emptive potentials.
Collapse
Affiliation(s)
- Saeed Mohammadi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, North Kargar Avenue, Tehran, 14117-13131 Iran
| | | | - Ashraf Malek Mohammadi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, North Kargar Avenue, Tehran, 14117-13131 Iran
| | - Hajar Nasiri
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, North Kargar Avenue, Tehran, 14117-13131 Iran
| | - Mohsen Nikbakht
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, North Kargar Avenue, Tehran, 14117-13131 Iran
| | - Najmaldin Saki
- Thalassemia and Hemoglobinopathy Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Vaezi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, North Kargar Avenue, Tehran, 14117-13131 Iran
| | - Kamran Alimoghaddam
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, North Kargar Avenue, Tehran, 14117-13131 Iran
| | - Ardeshir Ghavamzadeh
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, North Kargar Avenue, Tehran, 14117-13131 Iran
| |
Collapse
|
56
|
Wei YQ, Cen XN, Liu HH, Sun YH, Shi YJ, Liu W, Dong YJ, Ren HY. Irradiation of peripheral blood mononuclear cells with 7.5 Gy X-rays prior to donor lymphocyte infusion inhibits proliferation while preserving cytotoxicity, and improves the effectiveness of HSCT in patients with hematological malignancies. Oncol Lett 2017; 13:4101-4108. [PMID: 28588699 PMCID: PMC5452913 DOI: 10.3892/ol.2017.5966] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 02/03/2017] [Indexed: 12/02/2022] Open
Abstract
The aim of the present study was to explore the effect of different X-ray doses on the proliferation and cytotoxic activity of peripheral blood mononuclear cells (PBMCs), particularly lymphocytes, in order to assess whether this reduces the incidence of graft vs. host disease (GVHD) while preserving the graft vs. tumor (GVT) effect in patients with hematological malignancies following hematopoietic stem cell transplantation (HSCT). PBMCs from healthy donors were irradiated with X-rays at doses of 0, 2.5, 5, 7.5, 10, 15, 25 or 50 Gy, and their proliferative activity was determined using a WST-8 assay kit. The cytotoxic activity of non-irradiated PBMCs and PBMCs irradiated with 7.5 Gy X-rays was tested in the leukemic cell line K562 and its Adriamycin-resistant strain K562A using a lactate dehydrogenase assay. The clinical data of 7 patients who received 7.5 Gy X-ray-irradiated PBMC infusions following autologous HSCT were analyzed. PBMCs irradiated with ≥7.5 Gy X-rays exhibited a complete inhibition of proliferation. PBMCs irradiated with 7.5 Gy X-rays exhibited significantly increased cytotoxic activity towards K562 cells compared with K562A cells (P<0.05). There was no significant difference in cytotoxicity between irradiated and non-irradiated PBMCs, irrespective of the target cell, K562 or K562A (P>0.05). Based on the in vitro data, clinical data from patients who received 7.5 Gy X-ray-irradiated PBMC infusions following HSCT between January 2005 and January 2013 were assessed retrospectively. A total of 7 patients were included in the current study. The majority achieved various degrees of remission following donor lymphocyte infusion (DLI) and none suffered from GVHD. This indicates that 7.5 Gy-irradiated PMBCs have a potential application in DLI for the treatment of patients following HSCT. However, further studies on larger patient cohorts are required to assess the clinical potential of 7.5 Gy-irradiated PBMCs for preserving the GVT effect while avoiding GVHD following HSCT.
Collapse
Affiliation(s)
- Yong-Qiu Wei
- Department of Hematology, Peking University First Hospital, Beijing 100034, P.R. China
| | - Xi-Nan Cen
- Department of Hematology, Peking University First Hospital, Beijing 100034, P.R. China
| | - Hui-Hui Liu
- Department of Hematology, Peking University First Hospital, Beijing 100034, P.R. China
| | - Yu-Hua Sun
- Department of Hematology, Peking University First Hospital, Beijing 100034, P.R. China
| | - Yong-Jin Shi
- Department of Hematology, Peking University First Hospital, Beijing 100034, P.R. China
| | - Wei Liu
- Department of Hematology, Peking University First Hospital, Beijing 100034, P.R. China
| | - Yu-Jun Dong
- Department of Hematology, Peking University First Hospital, Beijing 100034, P.R. China
| | - Han-Yun Ren
- Department of Hematology, Peking University First Hospital, Beijing 100034, P.R. China
| |
Collapse
|
57
|
Miyamoto T, Fukuda T, Nakashima M, Henzan T, Kusakabe S, Kobayashi N, Sugita J, Mori T, Kurokawa M, Mori SI. Donor Lymphocyte Infusion for Relapsed Hematological Malignancies after Unrelated Allogeneic Bone Marrow Transplantation Facilitated by the Japan Marrow Donor Program. Biol Blood Marrow Transplant 2017; 23:938-944. [DOI: 10.1016/j.bbmt.2017.02.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 02/15/2017] [Indexed: 11/29/2022]
|
58
|
Saruta H, Ohata C, Muto I, Imamura T, Oku E, Ohshima K, Nagafuji K, Nakama T. Hematopoietic stem cell transplantation in advanced cutaneous T-cell lymphoma. J Dermatol 2017; 44:1038-1042. [PMID: 28391645 DOI: 10.1111/1346-8138.13848] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 02/19/2017] [Indexed: 11/30/2022]
Abstract
We retrospectively reviewed data pertaining to five patients with cutaneous T-cell lymphoma (CTCL) who had received hematopoietic stem cell transplantation (HSCT) between 2004 and 2015 at Kurume University Hospital, along with their clinical data until March 2016. For patients with advanced CTCL eligible for HSCT, autologous HSCT was performed when they responded well to chemotherapy, and allogeneic HSCT was selected for patients with advanced mycosis fungoides (MF)/Sézary syndrome (SS) and CTCL other than MF/SS with poor chemosensitivity. Two patients (primary cutaneous anaplastic large cell lymphoma and primary cutaneous CD8+ aggressive epidermotropic cytotoxic T-cell lymphoma) who responded well to chemotherapy received autologous HSCT: one patient was alive in partial remission and the other died due to therapy-related acute myeloid leukemia without disease relapse. In the remaining three patients with MF or SS, allogeneic HSCT was performed. Although one patient with MF died due to disease progression, the remaining two patients were alive in complete remission. Although there were two deaths in this study, the outcomes were considered satisfactory.
Collapse
Affiliation(s)
- Hiroshi Saruta
- Department of Dermatology, Kurume University School of Medicine, Kurume, Japan
| | - Chika Ohata
- Department of Dermatology, Kurume University School of Medicine, Kurume, Japan
| | - Ikko Muto
- Department of Dermatology, Kurume University School of Medicine, Kurume, Japan
| | - Taichi Imamura
- Department of Dermatology, Kurume University School of Medicine, Kurume, Japan
| | - Eijiro Oku
- Division of Hematology and Oncology, Department of Medicine, Kitakyushu General Hospital, Kitakyushu, Japan
| | - Koichi Ohshima
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Koji Nagafuji
- Division of Hematology and Oncology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Takekuni Nakama
- Department of Dermatology, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
59
|
Neuchel C, Fürst D, Niederwieser D, Bunjes D, Tsamadou C, Wulf G, Pfreundschuh M, Wagner E, Stuhler G, Einsele H, Schrezenmeier H, Mytilineos J. Impact of Donor Activating KIR Genes on HSCT Outcome in C1-Ligand Negative Myeloid Disease Patients Transplanted with Unrelated Donors-A Retrospective Study. PLoS One 2017; 12:e0169512. [PMID: 28107369 PMCID: PMC5249182 DOI: 10.1371/journal.pone.0169512] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 12/19/2016] [Indexed: 12/02/2022] Open
Abstract
Natural Killer cells (NK) are lymphocytes with the potential to recognize and lyse cells which escaped T-cell mediated lysis due to their aberrant HLA expression profiles. Killer cell immunoglobulin-like receptors (KIR) influence NK-cell activity by mediation of activating or inhibitory signals upon interaction with HLA-C (C1, C2) ligands. Therefore, absence of ligands for donor inhibitory KIRs following hematopoietic stem cell transplantation (HSCT) may have an influence on its outcome. Previous studies showed that C1 negative patients have a decreased HSCT outcome. Our study, based on a cohort of 200 C1-negative patients, confirmed these findings for the endpoints: overall survival (OS: HR = 1.41, CI = 1.14–1.74, p = 0.0012), disease free survival (DFS: HR = 1.27, CI = 1.05–1.53, p = 0.015), treatment related mortality (TRM: HR = 1.41, CI = 1.01–1.96, p = 0.04), and relapse incidence (RI: HR = 1.33, CI = 1.01–1.75, p = 0.04) all being inferior when compared to C1-positive patients (n = 1246). Subsequent analysis showed that these findings applied for patients with myeloid malignancies but not for patients with lymphoproliferative diseases (OS: myeloid: HR = 1.51, CI = 1.15–1.99, p = 0.003; lymphoblastic: HR = 1.26, CI = 0.91–1.75, p = 0.16; DFS: myeloid: HR = 1.31, CI = 1.01–1.70, p = 0.04; lymphoblastic: HR = 1.21, CI = 0.90–1.61, p = 0.21; RI: myeloid: HR = 1.31, CI = 1.01–1.70, p = 0.04; lymphoblastic: HR = 1.21, CI = 0.90–1.61, p = 0.21). Interestingly, within the C1-negative patient group, transplantation with KIR2DS2 resulted in better OS (9/10 matched: HR = 0.24, CI = 0.08–0.67, p = 0.007) as well as DFS (9/10 matched: HR = 0,26, CI = 0.11–0.60, p = 0.002), and transplantation with KIR2DS1 positive donors was associated with a decreased RI (HR = 0.30, CI = 0.13–0.69, p = 0.005). TRM was increased when the donor was positive for KIR2DS1 (HR = 2.61, CI = 1.26–5.41, p = 0.001). Our findings suggest that inclusion of KIR2DS1/2/5 and KIR3DS1-genotyping in the unrelated donor search algorithm of C1-ligand negative patients with myeloid malignancies may prove to be of clinical relevance.
Collapse
Affiliation(s)
- Christine Neuchel
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden Wuerttenberg–Hessen and University Hospital of Ulm, Ulm, Germany
- Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Daniel Fürst
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden Wuerttenberg–Hessen and University Hospital of Ulm, Ulm, Germany
- Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | | | - Donald Bunjes
- Department of Hematology/Oncology, University Clinic Ulm, Ulm, Germany
| | - Chrysanthi Tsamadou
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden Wuerttenberg–Hessen and University Hospital of Ulm, Ulm, Germany
- Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Gerald Wulf
- Department of Hematology/Oncology, Georg-August-University Göttingen, Göttingen, Germany
| | - Michael Pfreundschuh
- Department of Internal Medicine I, Universitätsklinikum des Saarlandes, Homburg, Germany
| | - Eva Wagner
- Department of Medicine III, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Gernot Stuhler
- Centre for Bone Marrow and Blood Stem Cell Transplantation, Deutsche Klinik für Diagnostik, Wiesbaden, Germany
| | - Hermann Einsele
- Department of Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Hubert Schrezenmeier
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden Wuerttenberg–Hessen and University Hospital of Ulm, Ulm, Germany
- Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Joannis Mytilineos
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden Wuerttenberg–Hessen and University Hospital of Ulm, Ulm, Germany
- Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
- DRST–German Registry for Stem Cell Transplantation, Essen, Germany
- * E-mail:
| |
Collapse
|
60
|
van Bergen CAM, van Luxemburg-Heijs SAP, de Wreede LC, Eefting M, von dem Borne PA, van Balen P, Heemskerk MHM, Mulder A, Claas FHJ, Navarrete MA, Honders WM, Rutten CE, Veelken H, Jedema I, Halkes CJM, Griffioen M, Falkenburg JHF. Selective graft-versus-leukemia depends on magnitude and diversity of the alloreactive T cell response. J Clin Invest 2017; 127:517-529. [PMID: 28067665 DOI: 10.1172/jci86175] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 11/17/2016] [Indexed: 01/10/2023] Open
Abstract
Patients with leukemia who receive a T cell-depleted allogeneic stem cell graft followed by postponed donor lymphocyte infusion (DLI) can experience graft-versus-leukemia (GVL) reactivity, with a lower risk of graft-versus-host disease (GVHD). Here, we have investigated the magnitude, diversity, and specificity of alloreactive CD8 T cells in patients who developed GVL reactivity after DLI in the absence or presence of GVHD. We observed a lower magnitude and diversity of CD8 T cells for minor histocompatibility antigens (MiHAs) in patients with selective GVL reactivity without GVHD. Furthermore, we demonstrated that MiHA-specific T cell clones from patients with selective GVL reactivity showed lower reactivity against nonhematopoietic cells, even when pretreated with inflammatory cytokines. Expression analysis of MiHA-encoding genes showed that similar types of antigens were recognized in both patient groups, but in patients who developed GVHD, T cell reactivity was skewed to target broadly expressed MiHAs. As an inflammatory environment can render nonhematopoietic cells susceptible to T cell recognition, prevention of such circumstances favors induction of selective GVL reactivity without development of GVHD.
Collapse
|
61
|
Baraban EG, Hu S, Hui P, Podoltsev N, Cooper D, Xu ML. Tissue-based chimerism analysis enhances detection of donor-derived neoplasia in allogeneic stem cell transplant patients. Bone Marrow Transplant 2016; 52:634-637. [PMID: 27991892 DOI: 10.1038/bmt.2016.332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- E G Baraban
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - S Hu
- Division of Pathology/Laboratory Medicine, Department of Hematopathology, University of Texas MD Anderson Cancer Center, Austin, TX, USA
| | - P Hui
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - N Podoltsev
- Department of Internal Medicine, Division of Hematology, Yale Cancer Center, Smilow Cancer Hospital at the Yale-New Haven Hospital, Yale University School of Medicine, New Haven, CT, USA
| | - D Cooper
- Blood and Marrow Transplant Program, Robert Wood Johnson University, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - M L Xu
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
62
|
Pre-Emptive Immunotherapy for Clearance of Molecular Disease in Childhood Acute Lymphoblastic Leukemia after Transplantation. Biol Blood Marrow Transplant 2016; 23:87-95. [PMID: 27742575 DOI: 10.1016/j.bbmt.2016.10.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 10/07/2016] [Indexed: 12/21/2022]
Abstract
Monitoring of minimal residual disease (MRD) or chimerism may help guide pre-emptive immunotherapy (IT) with a view to preventing relapse in childhood acute lymphoblastic leukemia (ALL) after transplantation. Patients with ALL who consecutively underwent transplantation in Frankfurt/Main, Germany between January 1, 2005 and July 1, 2014 were included in this retrospective study. Chimerism monitoring was performed in all, and MRD assessment was performed in 58 of 89 patients. IT was guided in 19 of 24 patients with mixed chimerism (MC) and MRD and by MRD only in another 4 patients with complete chimerism (CC). The 3-year probabilities of event-free survival (EFS) were .69 ± .06 for the cohort without IT and .69 ± .10 for IT patients. Incidences of relapse (CIR) and treatment-related mortality (CITRM) were equally distributed between both cohorts (without IT: 3-year CIR, .21 ± .05, 3-year CITRM, .10 ± .04; IT patients: 3-year CIR, .18 ± .09, 3-year CITRM .13 ± .07). Accordingly, 3-year EFS and 3-year CIR were similar in CC and MC patients with IT, whereas MC patients without IT experienced relapse. IT was neither associated with an enhanced immune recovery nor an increased risk for acute graft-versus-host disease. Relapse prevention by IT in patients at risk may lead to the same favorable outcome as found in CC and MRD-negative-patients. This underlines the importance of excellent MRD and chimerism monitoring after transplantation as the basis for IT to improve survival in childhood ALL.
Collapse
|
63
|
EXP CLIN TRANSPLANTExp Clin Transplant 2016; 14. [DOI: 10.6002/ect.2016.0074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
64
|
Berens C, Heine A, Müller J, Held SAE, Mayer K, Brossart P, Oldenburg J, Pötzsch B, Wolf D, Rühl H. Variable resistance to freezing and thawing of CD34-positive stem cells and lymphocyte subpopulations in leukapheresis products. Cytotherapy 2016; 18:1325-31. [PMID: 27491794 DOI: 10.1016/j.jcyt.2016.06.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 05/20/2016] [Accepted: 06/23/2016] [Indexed: 11/28/2022]
Abstract
BACKGROUND AIMS Leukapheresis products for hematopoietic stem cell transplantation can be cryopreserved for various indications. Although it is known that CD34(+) cells tolerate cryopreservation well, a significant loss of CD3(+) cells has been observed, which has been ascribed to several factors, including transport, storage conditions and granulocyte-colony stimulating factor (G-CSF) administration. METHODS To assess the tolerance of CD34(+) cells and lymphocyte subpopulations for cryopreservation and thawing, the post-thaw recoveries of CD34(+) cells, CD3(+)CD4(+) cells, CD3(+)CD8(+) cells, CD19(+) cells and CD16(+)CD56(+) cells were determined in 90 cryopreserved apheresis products, among which 65 were from G-CSF-mobilized donors, and 34 from unrelated donors that underwent transport before cryopreservation at our center. A controlled rate freezer and 5% dimethyl sulfoxide were used for cryopreservation. RESULTS We could detect statistically significant differences for CD34(+) cell recovery (93.0 ± 20.7%) when compared to CD3(+)CD4(+) cell (83.1 ± 15.4%, P = 0.014), and CD3(+)CD8(+) cell recovery (83.3 ± 13.9%, P = 0.001). Similarly, CD19(+) cell recovery (98.6 ± 15.1%) was higher than CD3(+)CD4(+) cell (P = 2.5 × 10(-7)) and CD3(+)CD8(+) cell recovery (P = 1.2 × 10(-8)). Post-thaw recovery rates of all cell populations were not impaired in G-CSF-mobilized products compared with non-mobilized products nor in unrelated compared with related donor products. DISCUSSION Our data suggest a lower tolerance of CD3(+) cells for cryopreservation and demonstrate that freezing-thawing resistance thawing is cell-specific and independent from other factors that affect post-thaw recovery of cryopreserved cells. Thus, a clinical consequence may be the monitoring of post-thaw CD3(+) cell doses of cryopreserved products, such as donor lymphocyte infusions.
Collapse
Affiliation(s)
- Christina Berens
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn (UKB), Bonn, Germany
| | - Annkristin Heine
- Department of Internal Medicine III, Oncology, Hematology and Rheumatology, University Hospital Bonn (UKB), Bonn, Germany
| | - Jens Müller
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn (UKB), Bonn, Germany
| | - Stefanie Andrea Erika Held
- Department of Internal Medicine III, Oncology, Hematology and Rheumatology, University Hospital Bonn (UKB), Bonn, Germany
| | - Karin Mayer
- Department of Internal Medicine III, Oncology, Hematology and Rheumatology, University Hospital Bonn (UKB), Bonn, Germany
| | - Peter Brossart
- Department of Internal Medicine III, Oncology, Hematology and Rheumatology, University Hospital Bonn (UKB), Bonn, Germany
| | - Johannes Oldenburg
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn (UKB), Bonn, Germany
| | - Bernd Pötzsch
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn (UKB), Bonn, Germany
| | - Dominik Wolf
- Department of Internal Medicine III, Oncology, Hematology and Rheumatology, University Hospital Bonn (UKB), Bonn, Germany
| | - Heiko Rühl
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn (UKB), Bonn, Germany.
| |
Collapse
|
65
|
Abstract
PURPOSE OF REVIEW Adoptive T-cell therapy has become one of the most exciting fields of cancer therapy in the past few years. In this article, we describe a method which combines adoptive T-cell therapy with antibody therapy by arming T cells from cord blood, normal patients, and cancer patients with bispecific antibodies capable of binding to tumor-associated antigens on one side of the bispecific antibody construct and T cells on another side of the construct. This approach redirects T cells against tumor cells in a non-MHC-restricted manner. RECENT FINDINGS Various methods for manipulating the immune system including check-point inhibitors, chimeric antigen receptor T cells, and bispecific antibodies have shown promising activity in treating both hematological malignancies and solid tumors with excellent success. In recent studies, activated T cells armed with bispecific antibodies have shown good preclinical activity, safety, and promising efficacy in the clinical trials. SUMMARY Activated T cells armed with bispecific antibodies represent a promising treatment for cancer immunotherapy.
Collapse
|
66
|
CAR T Cell Therapy: A Game Changer in Cancer Treatment. J Immunol Res 2016; 2016:5474602. [PMID: 27298832 PMCID: PMC4889848 DOI: 10.1155/2016/5474602] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 04/28/2016] [Accepted: 05/04/2016] [Indexed: 01/13/2023] Open
Abstract
The development of novel targeted therapies with acceptable safety profiles is critical to successful cancer outcomes with better survival rates. Immunotherapy offers promising opportunities with the potential to induce sustained remissions in patients with refractory disease. Recent dramatic clinical responses in trials with gene modified T cells expressing chimeric antigen receptors (CARs) in B-cell malignancies have generated great enthusiasm. This therapy might pave the way for a potential paradigm shift in the way we treat refractory or relapsed cancers. CARs are genetically engineered receptors that combine the specific binding domains from a tumor targeting antibody with T cell signaling domains to allow specifically targeted antibody redirected T cell activation. Despite current successes in hematological cancers, we are only in the beginning of exploring the powerful potential of CAR redirected T cells in the control and elimination of resistant, metastatic, or recurrent nonhematological cancers. This review discusses the application of the CAR T cell therapy, its challenges, and strategies for successful clinical and commercial translation.
Collapse
|
67
|
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: 51] [Impact Index Per Article: 5.7] [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
|
68
|
Admiraal R, Boelens JJ. Individualized conditioning regimes in cord blood transplantation: Towards improved and predictable safety and efficacy. Expert Opin Biol Ther 2016; 16:801-13. [DOI: 10.1517/14712598.2016.1164688] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
69
|
Griffioen M, van Bergen CAM, Falkenburg JHF. Autosomal Minor Histocompatibility Antigens: How Genetic Variants Create Diversity in Immune Targets. Front Immunol 2016; 7:100. [PMID: 27014279 PMCID: PMC4791598 DOI: 10.3389/fimmu.2016.00100] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 03/01/2016] [Indexed: 11/13/2022] Open
Abstract
Allogeneic stem cell transplantation (alloSCT) can be a curative treatment for hematological malignancies. Unfortunately, the desired anti-tumor or graft-versus-leukemia (GvL) effect is often accompanied with undesired side effects against healthy tissues known as graft-versus-host disease (GvHD). After HLA-matched alloSCT, GvL and GvHD are both mediated by donor-derived T-cells recognizing polymorphic peptides presented by HLA surface molecules on patient cells. These polymorphic peptides or minor histocompatibility antigens (MiHA) are produced by genetic differences between patient and donor. Since polymorphic peptides may be useful targets to manipulate the balance between GvL and GvHD, the dominant repertoire of MiHA needs to be discovered. In this review, the diversity of autosomal MiHA characterized thus far as well as the various molecular mechanisms by which genetic variants create immune targets and the role of cryptic transcripts and proteins as antigen sources are described. The tissue distribution of MiHA as important factor in GvL and GvHD is considered as well as possibilities how hematopoietic MiHA can be used for immunotherapy to augment GvL after alloSCT. Although more MiHA are still needed for comprehensive understanding of the biology of GvL and GvHD and manipulation by immunotherapy, this review shows insight into the composition and kinetics of in vivo immune responses with respect to specificity, diversity, and frequency of specific T-cells and surface expression of HLA-peptide complexes and other (accessory) molecules on the target cell. A complex interplay between these factors and their environment ultimately determines the spectrum of clinical manifestations caused by immune responses after alloSCT.
Collapse
Affiliation(s)
- Marieke Griffioen
- Department of Hematology, Leiden University Medical Center , Leiden , Netherlands
| | | | | |
Collapse
|
70
|
Orti G, Sanz J, Bermudez A, Caballero D, Martinez C, Sierra J, Cabrera Marin JR, Espigado I, Solano C, Ferrà C, García-Noblejas A, Jimenez S, Sampol A, Yañez L, García-Gutiérrez V, Pascual MJ, Jurado M, Moraleda JM, Valcarcel D, Sanz MA, Carreras E, Duarte RF. Outcome of Second Allogeneic Hematopoietic Cell Transplantation after Relapse of Myeloid Malignancies following Allogeneic Hematopoietic Cell Transplantation: A Retrospective Cohort on Behalf of the Grupo Español de Trasplante Hematopoyetico. Biol Blood Marrow Transplant 2016; 22:584-8. [DOI: 10.1016/j.bbmt.2015.11.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 11/16/2015] [Indexed: 11/26/2022]
|
71
|
Quality of life in chronic myeloid leukaemia patients after haematopoietic cell transplantation pretreated with second-generation tyrosine kinase inhibitors. Contemp Oncol (Pozn) 2016; 20:414-417. [PMID: 28373825 PMCID: PMC5371710 DOI: 10.5114/wo.2016.64607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 10/09/2015] [Indexed: 11/21/2022] Open
Abstract
Aim of the study The majority of patients with chronic myeloid leukaemia (CML) respond to tyrosine kinase inhibitors (TKI), while allogeneic haematopoietic cell transplantation (HCT) is indicated in selected clinical situations. HCT carries the risk of severe complications, while the toxicity profile of dasatinib and nilotinib may lead to adverse reactions affecting the quality of life (QoL). We present the results of observational analysis of CML patients who underwent HCT after exposure to second-generation TKI (TKI2), with respect to their quality of life assessed comparatively after transplantation. Material and methods Eligible subjects included 19 patients. The quality of life and global health assessment were performed with a questionnaire comparing the signs and symptoms present during the TKI2-therapy with those related to post-transplant complications, including psychosocial problems. Results and conclusions Most patients had no/few problems with exhausting activities, no/few difficulties during long-distance walks, and do not/rarely rest in the daytime. Seventeen (89.5%) patients reported at least one symptom related to TKI2-therapy and most of them disappeared after HCT. Thirteen (68.4%) patients noted no serious complication after HCT. Most patients claimed to have a very good QoL and general health compared to the period prior to HCT. We found statistically significant improvement in global health (p = 0.016) and QoL (p = 0.043) after HCT. From the survivors perspective, HCT influence positively general health and QoL comparing to TKI2-therapy period. Further studies on larger group of patients will more precisely define the QoL level and possible predictors of changes in QoL, to assess which group of patients needs psychological support.
Collapse
|
72
|
McLaughlin L, Cruz CR, Bollard CM. Adoptive T-cell therapies for refractory/relapsed leukemia and lymphoma: current strategies and recent advances. Ther Adv Hematol 2015; 6:295-307. [PMID: 26622998 DOI: 10.1177/2040620715594736] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Despite significant advancements in the treatment and outcome of hematologic malignancies, prognosis remains poor for patients who have relapsed or refractory disease. Adoptive T-cell immunotherapy offers novel therapeutics that attempt to utilize the noted graft versus leukemia effect. While CD19 chimeric antigen receptor (CAR)-modified T cells have thus far been the most clinically successful application of adoptive T immunotherapy, further work with antigen specific T cells and CARs that recognize other targets have helped diversify the field to treat a broad spectrum of hematologic malignancies. This article will focus primarily on therapies currently in the clinical trial phase as well as current downfalls or limitations.
Collapse
Affiliation(s)
- Lauren McLaughlin
- Children's National Health System and The George Washington University, Washington, DC, USA
| | - C Russell Cruz
- Children's National Health System and The George Washington University, Washington, DC, USA
| | - Catherine M Bollard
- Children's National Health System and The George Washington University, 111 Michigan Ave, Washington, DC 20010, USA
| |
Collapse
|
73
|
New strategies of DLI in the management of relapse of hematological malignancies after allogeneic hematopoietic SCT. Bone Marrow Transplant 2015; 51:324-32. [PMID: 26595077 DOI: 10.1038/bmt.2015.288] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 09/24/2015] [Accepted: 10/19/2015] [Indexed: 12/17/2022]
Abstract
DLI is an effective strategy for patients with recurrent hematological malignancies after allogeneic hematopoietic SCT (allo-HSCT). DLI has been widely applied to boost the graft vs tumor (GVT) or GVL effects. However, given the potentially severe complications associated with conventional DLI and transient GVL effect, new strategies for DLI are emerging. In this review, we have discussed the recent important studies on DLI as a prophylactic or therapeutic modality for relapsed hematological disorders after allo-HSCT. The strategies to separate GVL from GVHD have also been discussed. Leukemia-targeting therapy and lymphodepletion combined with DLI, and prophylactic DLI after allo-HSCT are often employed for patients with high risk of relapse, which has been reviewed as well. In addition, we have also discussed the issues on DLI to be further addressed, such as the doses, timing and frequency of DLI in different clinical settings, leukemic antigen-specific DLI as well as how to augment GVL effect while attenuating GVHD.
Collapse
|
74
|
Dai H, Zhang W, Li X, Han Q, Guo Y, Zhang Y, Wang Y, Wang C, Shi F, Zhang Y, Chen M, Feng K, Wang Q, Zhu H, Fu X, Li S, Han W. Tolerance and efficacy of autologous or donor-derived T cells expressing CD19 chimeric antigen receptors in adult B-ALL with extramedullary leukemia. Oncoimmunology 2015; 4:e1027469. [PMID: 26451310 PMCID: PMC4590028 DOI: 10.1080/2162402x.2015.1027469] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 03/01/2015] [Accepted: 03/03/2015] [Indexed: 01/16/2023] Open
Abstract
The engineering of T lymphocytes to express chimeric antigen receptors (CARs) aims to establish T cell-mediated tumor immunity rapidly. In this study, we conducted a pilot clinical trial of autologous or donor- derived T cells genetically modified to express a CAR targeting the B-cell antigen CD19 harboring 4-1BB and the CD3ζ moiety. All enrolled patients had relapsed or chemotherapy-refractory B-cell lineage acute lymphocytic leukemia (B-ALL). Of the nine patients, six had definite extramedullary involvement, and the rate of overall survival at 18 weeks was 56%. One of the two patients who received conditioning chemotherapy achieved a three-month durable complete response with partial regression of extramedullary lesions. Four of seven patients who did not receive conditioning chemotherapy achieved dramatic regression or a mixed response in the haematopoietic system and extramedullary tissues for two to nine months. Grade 2-3 graft-versus-host disease (GVHD) was observed in two patients who received substantial donor-derived anti-CD19 CART (chimeric antigen receptor-modified T) cells 3-4 weeks after cell infusions. These results show for the first time that donor-derived anti-CD19 CART cells can cause GVHD and regression of extramedullary B-ALL. This study is registered at www.clinicaltrials.gov as NCT01864889.
Collapse
Affiliation(s)
- Hanren Dai
- Department of Immunology; Institute of Basic Medicine; School of Life Sciences; Chinese PLA General Hospital; Beijing, China
| | - Wenying Zhang
- Department of Bio-therapeutic; Chinese PLA General Hospital; Beijing, China
| | - Xiaolei Li
- Department of Molecular Biology; Institute of Basic Medicine; School of Life Sciences; Chinese PLA General Hospital; Beijing, China
| | - Qingwang Han
- Department of Immunology; Institute of Basic Medicine; School of Life Sciences; Chinese PLA General Hospital; Beijing, China
| | - Yelei Guo
- Department of Immunology; Institute of Basic Medicine; School of Life Sciences; Chinese PLA General Hospital; Beijing, China
| | - Yajing Zhang
- Department of Bio-therapeutic; Chinese PLA General Hospital; Beijing, China
| | - Yao Wang
- Department of Immunology; Institute of Basic Medicine; School of Life Sciences; Chinese PLA General Hospital; Beijing, China
| | - Chunmeng Wang
- Department of Bio-therapeutic; Chinese PLA General Hospital; Beijing, China
| | - Fengxia Shi
- Department of Bio-therapeutic; Chinese PLA General Hospital; Beijing, China
| | - Yan Zhang
- Department of Bio-therapeutic; Chinese PLA General Hospital; Beijing, China
| | - Meixia Chen
- Department of Bio-therapeutic; Chinese PLA General Hospital; Beijing, China
| | - Kaichao Feng
- Department of Bio-therapeutic; Chinese PLA General Hospital; Beijing, China
| | - Quanshun Wang
- Department of Hematology; Chinese PLA General Hospital; Beijing, China
| | - Hongli Zhu
- Department of Hematology; Chinese PLA General Hospital; Beijing, China
| | - Xiaobing Fu
- Department of Bio-therapeutic; Chinese PLA General Hospital; Beijing, China
| | - Suxia Li
- Department of Hematology; Chinese PLA General Hospital; Beijing, China
| | - Weidong Han
- Department of Immunology; Institute of Basic Medicine; School of Life Sciences; Chinese PLA General Hospital; Beijing, China
- Department of Bio-therapeutic; Chinese PLA General Hospital; Beijing, China
- Department of Molecular Biology; Institute of Basic Medicine; School of Life Sciences; Chinese PLA General Hospital; Beijing, China
| |
Collapse
|
75
|
Pereira TDM, Danby R, Rocha V. Donor lymphocyte infusion after allogeneic hematopoietic stem cell transplantation. Int J Hematol Oncol 2015. [DOI: 10.2217/ijh.15.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Donor lymphocyte infusion, a rescue therapy after hematopoietic stem cell transplantation, has been increasingly adopted, as modalities of stem cell transplantation have widened. First described as donor lymphocyte transfusion or cell therapy, it consists of infusion of donor lymphocytes, collected in steady state or after growth factor enhancement. As in literature the most used name is donor lymphocyte infusion, we'll adopt it here. Its most striking efficacy is observed in patients with chronic myelogenous leukemia, who relapsed after allogeneic stem cells transplantation. However, graft-versus-host disease, its main complication, may still hamper its feasibility.
Collapse
Affiliation(s)
- Thales Dalessandro Meneguin Pereira
- Oxford University Hospitals NHS Trust, Department of Clinical Haematology, Level 2, Cancer & Haematology Centre, Churchill Hospital, Old Road, Headington, Oxford, OX3 7LE, UK
| | - Robert Danby
- Oxford University Hospitals NHS Trust, Department of Clinical Haematology, Level 2, Cancer & Haematology Centre, Churchill Hospital, Old Road, Headington, Oxford, OX3 7LE, UK
| | - Vanderson Rocha
- Oxford University Hospitals NHS Trust, Department of Clinical Haematology, Level 2, Cancer & Haematology Centre, Churchill Hospital, Old Road, Headington, Oxford, OX3 7LE, UK
- BRC Blood Theme, NIHR Oxford Biomedical Centre, Oxford University Hospital, Oxford, OX3 9DS, UK
| |
Collapse
|
76
|
Piekarska A, Gil L, Prejzner W, Wiśniewski P, Leszczyńska A, Gniot M, Komarnicki M, Hellmann A. Pretransplantation use of the second-generation tyrosine kinase inhibitors has no negative impact on the HCT outcome. Ann Hematol 2015. [PMID: 26220759 PMCID: PMC4569656 DOI: 10.1007/s00277-015-2457-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Introduction Allogeneic hematopoietic cell transplantation (HCT) was a standard therapy in chronic phase (CP) chronic myeloid leukemia (CML). As a result of the effective therapy with tyrosine kinase inhibitors (TKI), HCT was shifted to defined clinical situations. We present the results of observational prospective analysis of 28 CML patients undergoing HCT after exposure to, at least, two lines of TKI (including dasatinib and/or nilotinib), with respect to response, overall survival (OS), treatment toxicity, graft versus host disease (GVHD), and progression/relapse incidence. Results All the patients but one engrafted with median time 19 days. OS for patients in CP1 and CP2/accelerated phase (AcP) were 92.9 and 85.7 %, respectively. Six patients allotransplanted in blast crisis (BC) CML died early after HCT. Eighteen patients achieved deep molecular remission (MR4.5 or MR4.0). Relapse incidence was 29.6 %. Median time to progression (TTP) differs significantly depending on the CML phase prior to HCT, the best response achieved after HCT and development of chronic GvHD. NRM yielded the values 7.1, 12.5, and 50 % in CP1, CP2/AcP, and BC, respectively. Fatal outcome, due to veno-occlusive disease (VOD), was observed in two (7 %) patients. In five (17.9 %) patients, mild or moderate VOD was observed with no negative impact of preceding therapy with TKI2. Acute GvHD was diagnosed in 25.9 % of patients, while chronic GvHD developed in 42.9 % of individuals. Conclusion Pretransplantation therapy with TKI2 in CP CML is safe and reasonable. In BC, the optimal approach before HCT is to reduce the leukemic burden and achieve subsequent CP.
Collapse
Affiliation(s)
- Agnieszka Piekarska
- Department of Hematology and Transplantology, Medical University of Gdansk, Debinki 7, 80-952, Gdansk, Poland.
| | - Lidia Gil
- Department of Hematology and Bone Marrow Transplantation, Poznan University of Medical Sciences, Szamarzewskiego 84, 60-569, Poznan, Poland
| | - Witold Prejzner
- Department of Hematology and Transplantology, Medical University of Gdansk, Debinki 7, 80-952, Gdansk, Poland
| | - Piotr Wiśniewski
- Department of Endocrinology and Internal Diseases, Medical University of Gdansk, Debinki 7, 80-952, Gdansk, Poland
| | - Aleksandra Leszczyńska
- Department of Hematology and Transplantology, Medical University of Gdansk, Debinki 7, 80-952, Gdansk, Poland
| | - Michał Gniot
- Department of Hematology and Bone Marrow Transplantation, Poznan University of Medical Sciences, Szamarzewskiego 84, 60-569, Poznan, Poland
| | - Mieczysław Komarnicki
- Department of Hematology and Bone Marrow Transplantation, Poznan University of Medical Sciences, Szamarzewskiego 84, 60-569, Poznan, Poland
| | - Andrzej Hellmann
- Department of Hematology and Transplantology, Medical University of Gdansk, Debinki 7, 80-952, Gdansk, Poland
| |
Collapse
|
77
|
Xiong Y, Bensoussan D, Decot V. Adoptive Immunotherapies After Allogeneic Hematopoietic Stem Cell Transplantation in Patients With Hematologic Malignancies. Transfus Med Rev 2015; 29:259-67. [PMID: 26282736 DOI: 10.1016/j.tmrv.2015.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 07/01/2015] [Accepted: 07/05/2015] [Indexed: 12/25/2022]
Abstract
Hematopoietic stem cell transplantation (HSCT) is the only curative therapy for patients with chemotherapy-resistant hematologic malignancies that are usually fatal in absence of treatment. Hematopoietic stem cell transplantation is associated with significant early and late morbidity and mortality. Graft-versus-host disease, infections, and relapse are the most important causes of mortality after HSCT. Until now, these complications have been managed mainly with pharmacological drugs, but in some situations, this approach clearly shows its limit. As such, there is a significant need for novel therapies for the treatment of complications after allogeneic HSCT. In this review, the currently available adoptive immunotherapies offering an alternative in case of treatment failure of HSCT complications will be described. The results of the main clinical trials based on immune cell infusion will be discussed and the strategies aiming at maximizing cytotoxic T-lymphocyte, regulatory T-cell, natural killer cell, cytokine-induced killer cell, and γδ T-cell efficacies in the context of immunotherapy approaches after allogeneic HSCT in patients with hematologic malignancies will be gathered.
Collapse
Affiliation(s)
- Yu Xiong
- Unité de Thérapie Cellulaire et Tissus, CHU Nancy, Nancy, France; Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Danièle Bensoussan
- Unité de Thérapie Cellulaire et Tissus, CHU Nancy, Nancy, France; Nancy Université, UL-UMR CNRS 7365, Faculté de Médecine, Vandoeuvre-lés-Nancy, Nancy, France
| | - Véronique Decot
- Unité de Thérapie Cellulaire et Tissus, CHU Nancy, Nancy, France; Nancy Université, UL-UMR CNRS 7365, Faculté de Médecine, Vandoeuvre-lés-Nancy, Nancy, France.
| |
Collapse
|
78
|
Pico de Coaña Y, Choudhury A, Kiessling R. Checkpoint blockade for cancer therapy: revitalizing a suppressed immune system. Trends Mol Med 2015; 21:482-91. [PMID: 26091825 DOI: 10.1016/j.molmed.2015.05.005] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 05/19/2015] [Accepted: 05/20/2015] [Indexed: 01/22/2023]
Abstract
Immune checkpoint receptors are crucial molecules for fine-tuning immune responses. Checkpoint signaling dampens T cell activation to avoid autoimmunity and the destructive effects of an excessive inflammatory response. It is well established that tumors use several mechanisms to avoid elimination by the immune system, and one involves hijacking these checkpoint pathways. Checkpoint blockade therapy utilizes monoclonal antibodies to release the brakes from suppressed T cells, allowing them to be activated and recover their antitumor activity. This therapeutic approach has revolutionized cancer immunotherapy, and extraordinary increases in overall survival were noted, first with anti-CTLA-4 (cytotoxic T lymphocyte-associated protein 4) and subsequently with anti-PD-1 (programmed cell death receptor-1) in melanoma and other malignancies.
Collapse
Affiliation(s)
- Yago Pico de Coaña
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden.
| | | | - Rolf Kiessling
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
79
|
Klaver Y, Kunert A, Sleijfer S, Debets R, Lamers CHJ. Adoptive T-cell therapy: a need for standard immune monitoring. Immunotherapy 2015; 7:513-33. [DOI: 10.2217/imt.15.23] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cancer immune therapy, in particular the use of checkpoint inhibitors and adoptive transfer of T cells has recently demonstrated significant clinical responses against several tumor types. Unfortunately, these therapies are frequently accompanied by severe toxicities, underscoring the need for markers that provide information on therapy response. Monitoring immune responses in the tumor microenvironment and peripheral blood prior to and during these therapies will provide better insight into the mechanisms underlying clinical activities, and will potentially enable the identification of such markers. In this review, we present an overview of adoptive T-cell trials conducted with a special focus on immune monitoring, and argue that accurate monitoring of T cells is pivotal to further development of immune therapies to treat cancer.
Collapse
Affiliation(s)
- Yarne Klaver
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Andre Kunert
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Stefan Sleijfer
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Reno Debets
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Cor HJ Lamers
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
80
|
Infusion of donor lymphocytes expressing the herpes simplex virus thymidine kinase suicide gene for recurrent hematologic malignancies after allogeneic hematopoietic stem cell transplantation. Int J Hematol 2015; 102:101-10. [PMID: 25948083 DOI: 10.1007/s12185-015-1801-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 04/11/2015] [Accepted: 04/14/2015] [Indexed: 10/23/2022]
Abstract
The infusion of donor lymphocytes expressing the herpes simplex virus thymidine kinase suicide gene (TK-cells) is a promising strategy for the treatment of hematologic malignancies relapsing after allogeneic hematopoietic stem cell transplantation. Here we report the results of a phase I clinical trial designed to examine the feasibility, safety, and efficacy of donor lymphocyte infusion (DLI) of TK-cells. Three patients (two with malignant lymphomas, one with acute myeloid leukemia) were enrolled in the trial and received a single DLI of 1 × 10(7) or 5 × 10(7) TK-cells/kg. No local or systemic toxicity related to the gene-transfer procedure was observed. Two patients achieved stable disease. No patient had severe graft-versus-host disease requiring systemic steroid and/or ganciclovir administration. TK-cells were detected in the peripheral blood of all three patients by PCR, but did not persist longer than 28 days. Analysis of cytotoxic T lymphocyte activity detected no immune response against TK-cells by the recipient's own T cells. Flow cytometric analysis showed low proliferative activity and cytotoxic function of TK-cells. In conclusion, DLI of TK-cells was safely performed in all three patients. Our analysis suggests the probable cause of rapid disappearance of TK-cells to be insufficient in vivo expansion of TK-cells in these patients.
Collapse
|
81
|
Radujkovic A, Guglielmi C, Bergantini S, Iacobelli S, van Biezen A, Milojkovic D, Gratwohl A, Schattenberg AVMB, Verdonck LF, Niederwieser DW, de Witte T, Kröger N, Olavarria E. Donor Lymphocyte Infusions for Chronic Myeloid Leukemia Relapsing after Allogeneic Stem Cell Transplantation: May We Predict Graft-versus-Leukemia Without Graft-versus-Host Disease? Biol Blood Marrow Transplant 2015; 21:1230-6. [PMID: 25797175 DOI: 10.1016/j.bbmt.2015.03.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 03/13/2015] [Indexed: 01/01/2023]
Abstract
Donor lymphocyte infusions (DLI) are an effective treatment for relapsed chronic myeloid leukemia (CML) after allogeneic stem cell transplantation (alloSCT). Leukemia resistance and secondary graft-versus-host disease (GVHD) are major obstacles to success with DLI. The aim of this study was to identify pre-DLI factors associated with prolonged survival in remission without secondary GVHD. We retrospectively analyzed 500 patients treated with DLI for CML relapse (16% molecular, 30% cytogenetic, and 54% hematological) after alloSCT. The overall probabilities of failure- and secondary GVHD-free survival (FGFS) were 29% and 27% at 5 and 10 years after DLI, respectively. The type of relapse was the major factor influencing FGFS (40% for molecular and/or cytogenetic relapse and 20% for hematological relapse at 5 years, P < .001). Chronic GVHD before DLI and an interval <1 year between alloSCT and first DLI were independently associated with inferior FGFS in patients with molecular and/or cytogenetic relapse. Consequently, FGFS was 13%, 35%, to 56% at 5 years in patients with 2, 1, and 0 adverse features, respectively. In patients with hematological relapse, independent adverse prognostic factors for FGFS were initial dose of CD3(+) cells ≥ 50 × 10(6)/kg, donor-recipient sex mismatch, and chronic GVHD before DLI. FGFS was 0%, 17%, 33%, to 37% in patients with 3, 2, 1, and 0 adverse features, respectively. The probability of survival in remission without secondary GVHD was highest (>50% at 5 years) when DLI were given beyond 1 year from alloSCT for molecular and/or cytogenetic CML relapse that was not preceded by chronic GVHD.
Collapse
Affiliation(s)
- Aleksandar Radujkovic
- Department of Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany.
| | - Cesare Guglielmi
- Dipartimento di Medicina Clinica e Molecolare, Università "Sapienza", Rome, Italy
| | - Stefania Bergantini
- Dipartimento di Medicina Clinica e Molecolare, Università "Sapienza", Rome, Italy
| | - Simona Iacobelli
- University of Rome Tor Vergata, Centro Interdipartimentale di Biostatistica e Bioinformatica (CIBB), Rome, Italy
| | - Anja van Biezen
- Department of Medical Statistics and Bioinformatics, Leiden University, Leiden, The Netherlands
| | - Dragana Milojkovic
- Department of Haematology, Hammersmith Hospitals Trust, Imperial College London, London, United Kingdom
| | - Alois Gratwohl
- Department of Hematology, University Hospital Basel, Basel, Switzerland
| | | | - Leo F Verdonck
- Department of Haematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Theo de Witte
- Department of Tumor Immunology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | |
Collapse
|
82
|
Chimerism analysis in peripheral blood using indel quantitative real-time PCR is a useful tool to predict post-transplant relapse in acute leukemia. Bone Marrow Transplant 2014; 50:259-65. [DOI: 10.1038/bmt.2014.254] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 09/03/2014] [Accepted: 09/04/2014] [Indexed: 01/11/2023]
|
83
|
A multicentre UK study of GVHD following DLI: rates of GVHD are high but mortality from GVHD is infrequent. Bone Marrow Transplant 2014; 50:62-7. [PMID: 25310308 DOI: 10.1038/bmt.2014.227] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 08/26/2014] [Accepted: 08/28/2014] [Indexed: 12/25/2022]
Abstract
DLIs are frequently used following haematopoietic SCT (HSCT) in patients with risk of relapse but data on GVHD following DLI are scarce. We report on 68 patients who received DLI following HSCT. Most patients developed GVHD following DLI (71%), which was acute in 22 patients (32%) almost half of whom had grade III-IV acute GVHD (aGVHD). Thirty patients (44%) developed cGVHD which followed aGVHD in four patients and was graded severe in nine patients. Corticosteroids were the most common first-line therapy for both acute and chronic GVHD. A wide range of second/third-line agents included cyclosporin, mycophenolate, tacrolimus, imatinib, infliximab and ECP. Relapse of initial malignancy occurred in 37%. Relapse was significantly less frequent in those receiving pre-emptive DLI. Relapse rates were also lower in those with GVHD (31%) than those without GVHD (50%), but this did not reach statistical significance. At 55 months post DLI, 34% of patients had died most commonly from relapse and 22% had on-going GVHD. Although GVHD was an important cause of morbidity post DLI (71%), only 6% died from GVHD. Although most patients develop GVHD post DLI and may require consecutive therapies, mortality from GVHD is infrequent. DLI remains an important option for relapse post transplant and manipulation of the GVT effect needs to be optimised to induce remission without morbidity from GVHD.
Collapse
|
84
|
Adoptive therapy with donor lymphocyte infusion after allogenic hematopoietic SCT in pediatric patients. Bone Marrow Transplant 2014; 50:51-5. [PMID: 25310307 DOI: 10.1038/bmt.2014.200] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 06/23/2014] [Accepted: 07/23/2014] [Indexed: 01/14/2023]
Abstract
The aim of this study was to analyse the experience of Polish Pediatric Group for Hematopoietic Stem Cell Transplantation in respect to donor lymphocyte infusion procedure. The study included 51 pediatric patients with malignant (45) and non-malignant (6) diseases treated with DLI in the period 1993-2012. The indications for DLI were as follows: (1) increasing recipient chimerism after non-ablative hematopoietic SCT (18 patients); (2) immunomodulation after a reduced intensity conditioning regimen (2 patients); (3) increase in minimal residual disease detection (3 patients); and (4) relapse (28 patients). DLI was carried out at a median of 6 (0.5-79) months after SCT. DLI was administered as either a single-dose (in 19 cases) or in escalating-dose regimens (in 32 cases). The median total dose of CD3-positive T cells was 28.0 (0.1-730.0) × 10(6)/kg body weight. The time for assessment of DLI efficacy ranged from 0 to 70 (median 3) months. At evaluation, 18 patients experienced CR, 3 achieved PR, 19 showed relapse and 11 rejected the graft. DLI was found to be effective in 39% of cases. Complications of the procedure occurred in 18 patients; of these, 2 died. To sum up DLI shows efficacy in a significant percentage of children. Mortality related to the therapy adverse effects is low. However, this method requires standardization.
Collapse
|
85
|
Qin XY, Li GX, Qin YZ, Wang Y, Wang FR, Liu DH, Xu LP, Chen H, Han W, Wang JZ, Zhang XH, Li JL, Li LD, Liu KY, Huang XJ. Quantitative chimerism: an independent acute leukemia prognosis indicator following allogeneic hematopoietic SCT. Bone Marrow Transplant 2014; 49:1269-77. [DOI: 10.1038/bmt.2014.158] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 05/26/2014] [Accepted: 05/30/2014] [Indexed: 11/09/2022]
|
86
|
Recommandations de la SFGM-TC concernant l’injection prophylactique, préemptive et curative des lymphocytes du donneur (DLI) après allogreffe de cellules souches hématopoïétiques. ACTA ACUST UNITED AC 2014; 62:193-6. [DOI: 10.1016/j.patbio.2014.05.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Accepted: 05/14/2014] [Indexed: 11/20/2022]
|
87
|
Rambaldi A, Biagi E, Bonini C, Biondi A, Introna M. Cell-based strategies to manage leukemia relapse: efficacy and feasibility of immunotherapy approaches. Leukemia 2014; 29:1-10. [PMID: 24919807 DOI: 10.1038/leu.2014.189] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 05/15/2014] [Accepted: 05/27/2014] [Indexed: 12/19/2022]
Abstract
When treatment fails, the clinical outcome of acute leukemia patients is usually very poor, particularly when failure occurs after transplantation. A second allogeneic stem cell transplant could be envisaged as an effective and feasible salvage option in younger patients having a late relapse and an available donor. Unmanipulated or minimally manipulated donor T cells may also be effective in a minority of patients but the main limit remains the induction of severe graft-versus-host disease. This clinical complication has brought about a huge research effort that led to the development of leukemia-specific T-cell therapy aiming at the direct recognition of leukemia-specific rather than minor histocompatibility antigens. Despite a great scientific interest, the clinical feasibility of such an approach has proven to be quite problematic. To overcome this limitation, more research has moved toward the choice of targeting commonly expressed hematopoietic specific antigens by the genetic modification of unselected T cells. The best example of this is represented by the anti-CD19 chimeric antigen receptor (CD19.CAR) T cells. As a possible alternative to the genetic manipulation of unselected T cells, specific T-cell subpopulations with in vivo favorable homing and long-term survival properties have been genetically modified by CAR molecules. Finally, the use of naturally cytotoxic effector cells such as natural killer and cytokine-induced killer cells has been proposed in several clinical trials. The clinical development of these latter cells could also be further expanded by additional genetic modifications using the CAR technology.
Collapse
Affiliation(s)
- A Rambaldi
- Hematology and Bone Marrow Transplant Unit and Center of Cell Therapy 'G. Lanzani', Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - E Biagi
- Department of Pediatrics, M Tettamanti Research Center, Laboratory of Cell therapy 'S. Verri' University of Milano-Bicocca, San Gerardo Hospital, Monza, Italy
| | - C Bonini
- Experimental Hematology Unit, San Raffaele Scientific Institute, Milano, Italy
| | - A Biondi
- Department of Pediatrics, M Tettamanti Research Center, Laboratory of Cell therapy 'S. Verri' University of Milano-Bicocca, San Gerardo Hospital, Monza, Italy
| | - M Introna
- Hematology and Bone Marrow Transplant Unit and Center of Cell Therapy 'G. Lanzani', Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| |
Collapse
|
88
|
Tan Y, Du K, Luo Y, Shi J, Cao L, Zheng Y, Zheng G, Zhao Y, Ye X, Cai Z, Huang H. Superiority of preemptive donor lymphocyte infusion based on minimal residual disease in acute leukemia patients after allogeneic hematopoietic stem cell transplantation. Transfusion 2013; 54:1493-500. [PMID: 24372162 DOI: 10.1111/trf.12524] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 09/25/2013] [Accepted: 10/01/2013] [Indexed: 12/30/2022]
Affiliation(s)
- Yamin Tan
- Bone Marrow Transplantation Center; The First Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou China
| | - Kaili Du
- Bone Marrow Transplantation Center; The First Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou China
| | - Yi Luo
- Bone Marrow Transplantation Center; The First Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou China
| | - Jimin Shi
- Bone Marrow Transplantation Center; The First Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou China
| | - Ling Cao
- Bone Marrow Transplantation Center; The First Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou China
| | - Yanlong Zheng
- Bone Marrow Transplantation Center; The First Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou China
| | - Gaofeng Zheng
- Bone Marrow Transplantation Center; The First Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou China
| | - Yanmin Zhao
- Bone Marrow Transplantation Center; The First Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou China
| | - Xiujing Ye
- Bone Marrow Transplantation Center; The First Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou China
| | - Zhen Cai
- Bone Marrow Transplantation Center; The First Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou China
| | - He Huang
- Bone Marrow Transplantation Center; The First Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou China
| |
Collapse
|
89
|
Cheadle EJ, Gornall H, Baldan V, Hanson V, Hawkins RE, Gilham DE. CAR T cells: driving the road from the laboratory to the clinic. Immunol Rev 2013; 257:91-106. [DOI: 10.1111/imr.12126] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Eleanor J. Cheadle
- Clinical and Experimental Immunotherapy Group; Department of Medical Oncology; Institute of Cancer Sciences; The University of Manchester; Manchester Academic Healthcare Science Centre; Manchester UK
- Targeted Therapy Group; Institute of Cancer Sciences; The University of Manchester; Manchester Academic Healthcare Science Centre; Manchester UK
| | - Hannah Gornall
- Clinical and Experimental Immunotherapy Group; Department of Medical Oncology; Institute of Cancer Sciences; The University of Manchester; Manchester Academic Healthcare Science Centre; Manchester UK
| | - Vania Baldan
- Clinical and Experimental Immunotherapy Group; Department of Medical Oncology; Institute of Cancer Sciences; The University of Manchester; Manchester Academic Healthcare Science Centre; Manchester UK
| | - Vivien Hanson
- Transplantation Laboratory; Oxford University Hospitals NHS Foundation Trust; Oxford UK
| | - Robert E. Hawkins
- Clinical and Experimental Immunotherapy Group; Department of Medical Oncology; Institute of Cancer Sciences; The University of Manchester; Manchester Academic Healthcare Science Centre; Manchester UK
| | - David E. Gilham
- Clinical and Experimental Immunotherapy Group; Department of Medical Oncology; Institute of Cancer Sciences; The University of Manchester; Manchester Academic Healthcare Science Centre; Manchester UK
| |
Collapse
|
90
|
Kochenderfer JN, Dudley ME, Carpenter RO, Kassim SH, Rose JJ, Telford WG, Hakim FT, Halverson DC, Fowler DH, Hardy NM, Mato AR, Hickstein DD, Gea-Banacloche JC, Pavletic SZ, Sportes C, Maric I, Feldman SA, Hansen BG, Wilder JS, Blacklock-Schuver B, Jena B, Bishop MR, Gress RE, Rosenberg SA. Donor-derived CD19-targeted T cells cause regression of malignancy persisting after allogeneic hematopoietic stem cell transplantation. Blood 2013; 122:4129-39. [PMID: 24055823 PMCID: PMC3862276 DOI: 10.1182/blood-2013-08-519413] [Citation(s) in RCA: 482] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 09/16/2013] [Indexed: 12/16/2022] Open
Abstract
New treatments are needed for B-cell malignancies persisting after allogeneic hematopoietic stem cell transplantation (alloHSCT). We conducted a clinical trial of allogeneic T cells genetically modified to express a chimeric antigen receptor (CAR) targeting the B-cell antigen CD19. T cells for genetic modification were obtained from each patient's alloHSCT donor. All patients had malignancy that persisted after alloHSCT and standard donor lymphocyte infusions (DLIs). Patients did not receive chemotherapy prior to the CAR T-cell infusions and were not lymphocyte depleted at the time of the infusions. The 10 treated patients received a single infusion of allogeneic anti-CD19-CAR T cells. Three patients had regressions of their malignancies. One patient with chronic lymphocytic leukemia (CLL) obtained an ongoing complete remission after treatment with allogeneic anti-CD19-CAR T cells, another CLL patient had tumor lysis syndrome as his leukemia dramatically regressed, and a patient with mantle cell lymphoma obtained an ongoing partial remission. None of the 10 patients developed graft-versus-host disease (GVHD). Toxicities included transient hypotension and fever. We detected cells containing the anti-CD19-CAR gene in the blood of 8 of 10 patients. These results show for the first time that donor-derived allogeneic anti-CD19-CAR T cells can cause regression of B-cell malignancies resistant to standard DLIs without causing GVHD.
Collapse
|
91
|
Fisher V, Khuu H, David-Ocampo V, Byrne K, Pavletic S, Bishop M, Fowler DH, Barrett AJ, Stroncek DF. Analysis of the recovery of cryopreserved and thawed CD34+ and CD3+ cells collected for hematopoietic transplantation. Transfusion 2013; 54:1088-92. [PMID: 24117879 DOI: 10.1111/trf.12428] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 07/26/2013] [Accepted: 07/30/2013] [Indexed: 01/24/2023]
Abstract
BACKGROUND Cryopreservation is often used to store cellular therapies, but little is known about how well CD3+ or CD34+ cells tolerate this process. STUDY DESIGN AND METHODS Viable CD34+ cell recoveries were analyzed from related and unrelated donor granulocyte-colony-stimulating factor (G-CSF)-mobilized peripheral blood stem cell (PBSC) products and viable CD3+ cell recoveries from G-CSF-mobilized and nonmobilized apheresis products from related and unrelated donors. All products were cryopreserved with 5% dimethyl sulfoxide and 6% pentastarch using a controlled-rate freezer and were stored in liquid nitrogen. Related donor products were cryopreserved immediately after collection and unrelated donor products greater than 12 hours postcollection. RESULTS The postthaw recovery of CD34+ cells from related donor PBSCs was high (n = 86; 97.5 ± 23.1%) and there was no difference in postthaw CD34+ cell recovery from unrelated donor PBSCs (n = 14; 98.8 ± 37.2%; p = 0.863). In related donor lymphocyte products the postthaw CD3+ cell recovery (n = 48; 90.7 ± 21.4%) was greater than that of unrelated donor products (n = 14; 66.6 ± 35.8%; p = 0.00251). All unrelated donor lymphocyte products were from G-CSF-mobilized products, while most related donor lymphocyte products were from nonmobilized products. A comparison of the CD3+ cell recovery from related donor G-CSF-mobilized products (n = 19; 85.0 ± 29.2%) with that of unrelated donor products found no significant difference (p = 0.137). CONCLUSIONS The postthaw recovery of CD34+ cells was high in both related and unrelated donor products, but the recovery of CD3+ cells in unrelated donor G-CSF-mobilized products was lower. G-CSF-mobilized unrelated donor products may contain fewer CD3+ cells than non-G-CSF-exposed products upon thaw and, when indicated, cell doses should be monitored.
Collapse
Affiliation(s)
- Virginia Fisher
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health (NIH), Bethesda, Maryland
| | | | | | | | | | | | | | | | | |
Collapse
|
92
|
de Lima M, Porter DL, Battiwalla M, Bishop MR, Giralt SA, Hardy NM, Kröger N, Wayne AS, Schmid C. Proceedings from the National Cancer Institute's Second International Workshop on the Biology, Prevention, and Treatment of Relapse After Hematopoietic Stem Cell Transplantation: part III. Prevention and treatment of relapse after allogeneic transplantation. Biol Blood Marrow Transplant 2013; 20:4-13. [PMID: 24018392 DOI: 10.1016/j.bbmt.2013.08.012] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Accepted: 08/30/2013] [Indexed: 12/16/2022]
Abstract
In the Second Annual National Cancer Institute's Workshop on the Biology, Prevention, and Treatment of Relapse after Hematopoietic Stem Cell Transplantation, the Scientific/Educational Session on the Prevention and Treatment of Relapse after Allogeneic Transplantation highlighted progress in developing new therapeutic approaches since the first relapse workshop. Recent insights that might provide a basis for the development of novel, practical clinical trials were emphasized, including utilization of newer agents, optimization of donor lymphocyte infusion (DLI), and investigation of novel cellular therapies. Dr. de Lima discussed pre-emptive and maintenance strategies to prevent relapse after transplantation, for example, recent promising results suggestive of enhanced graft-versus-tumor activity with hypomethylating agents. Dr. Schmid provided an overview of adjunctive strategies to improve cell therapy for relapse, including cytoreduction before DLI, combination of targeted agents with DLI, and considerations in use of second transplantations. Dr. Porter addressed strategies to enhance T cell function, including ex vivo activated T cells and T cell engineering, and immunomodulatory approaches to enhance T cell function in vivo, including exogenous cytokines and modulation of costimulatory pathways.
Collapse
Affiliation(s)
- Marcos de Lima
- Hematologic Malignancies and Stem Cell Transplant Program, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - David L Porter
- Blood and Marrow Transplantation, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Minoo Battiwalla
- Hematology Branch, National Heart, Lung and Blood Institute, Bethesda, Maryland
| | - Michael R Bishop
- Section of Hematology/Oncology, University of Chicago Medicine, Chicago, Illinois
| | - Sergio A Giralt
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Nancy M Hardy
- Experimental Transplantation Immunology Branch, National Cancer Institute, Center for Cancer Research, Bethesda, Maryland.
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alan S Wayne
- Pediatric Oncology Branch, National Cancer Institute, Center for Cancer Research, Bethesda, Maryland; Current: Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California
| | | |
Collapse
|
93
|
Risk-adapted donor lymphocyte infusion based on chimerism and donor source in pediatric leukemia. Blood Cancer J 2013; 3:e137. [PMID: 23995046 PMCID: PMC3763390 DOI: 10.1038/bcj.2013.39] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 07/23/2013] [Indexed: 11/08/2022] Open
Abstract
Donor lymphocyte infusion (DLI) is commonly used to treat leukemia relapse following stem cell transplantation. In florid relapse, however, the efficacy of DLI is limited with substantial risk of severe graft-versus-host disease (GvHD). Here, we develop a novel risk-adapted strategy characterized by pre-emptive DLI initiated at the time of mixed chimerism, a small starting dose based on donor source, dose-escalation guided by real-time chimerism monitoring and withholding of DLI immediately in patients achieving full donor chimerism. A total of 178 DLIs were given to 38 patients with mixed chimerism; thereafter, 33 patients (86.8%) had donor chimerism successfully increased, including 30 (78.9%) who had chimerism fully converted back to 100% donor. Cumulative incidence of relapse was significantly lower (P=0.00004) and overall survival higher (P=0.0003) in patients with chimerism fully corrected as compared with those of patients whose chimerism remained mixed. Only 13.2% of the patients developed acute grade III-IV GvHD with no associated mortality. In conclusion, the risk-adapted DLI strategy is useful in minimizing the risk of childhood leukemia relapse, GvHD and death.
Collapse
|
94
|
Stevanović S, Nijmeijer BA, van Schie ML, Salvatori DC, Maas S, Griffioen M, Falkenburg JF. Donor T Cells Administered Over HLA Class II Barriers Mediate Antitumor Immunity without Broad Off-Target Toxicity in a NOD/Scid Mouse Model of Acute Leukemia. Biol Blood Marrow Transplant 2013; 19:867-75. [DOI: 10.1016/j.bbmt.2013.03.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 03/06/2013] [Indexed: 01/21/2023]
|
95
|
Donor lymphocyte infusion for relapsed hematological malignancies after allogeneic hematopoietic cell transplantation: prognostic relevance of the initial CD3+ T cell dose. Biol Blood Marrow Transplant 2013; 19:949-57. [PMID: 23523892 DOI: 10.1016/j.bbmt.2013.03.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 03/06/2013] [Indexed: 11/20/2022]
Abstract
The impact of donor lymphocyte infusion (DLI) initial cell dose on its outcome is known in patients with chronic myeloid leukemia but limited in patients with other hematological malignancies. In this retrospective study, we evaluated the effect of initial DLI CD3(+) cell dose on graft-versus-host disease (GVHD) and overall survival after DLI given for relapse of any hematological malignancies after allogeneic hematopoietic cell transplantation (HCT) with high- or reduced-intensity conditioning. The cohort included 225 patients. Initial DLI CD3(+) cell dose per kilogram of recipient body weight was ≤ 1 × 10(7) (n = 84; group A), >1.0 to <10 × 10(7) (n = 58; group B), and ≥ 10 × 10(7) (n = 66; group C). The initial cell dose was unknown for the remaining 17 patients. Cumulative incidence rates of GVHD at 12 months after DLI were 21%, 45%, and 55% for groups A, B, and C, respectively. Multivariate analysis showed that initial DLI CD3(+) cell ≥ 10 × 10(7) dose per kilogram is associated with an increased risk of GVHD after DLI (P = .03). Moreover, an initial DLI CD3(+) cell dose of 10 × 10(7) or higher did not decrease the risk of relapse and did not improve overall survival. Thus, these results support the use of less than 10 × 10(7) CD3(+) cell per kilogram as the initial cell dose of DLI for treatment of persistent or recurrent hematological malignancy after HCT.
Collapse
|
96
|
Dalle JH. [Hematopoietic stem cell transplantation in 2012: who? Where? How?]. Arch Pediatr 2013; 20:405-11. [PMID: 23466405 DOI: 10.1016/j.arcped.2013.01.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Revised: 11/16/2012] [Accepted: 01/21/2013] [Indexed: 11/17/2022]
Abstract
Allogeneic bone marrow transplantation has dramatically changed over the years since its beginnings. The diseases treated with transplantation (malignant hemopathies, severe benign hemopathies such as congenital or acquired congenital medullary aplasia, hemoglobinopathies, as well as severe immune system deficiencies and certain overload diseases), stem cell sources (bone marrow, peripheral stem cells, placental blood), donor types (intrafamilial, nonrelated, totally or partially compatible), conditioning regimen (immunosuppressors, graft manipulation), and supportive care increasingly vary. Allogeneic stem cell transplantation and more widely cellular therapies now need to be discussed. In this paper, we propose an overview of these therapies in 2012 for pediatric patients.
Collapse
Affiliation(s)
- J-H Dalle
- Service d'hématologie et immunologie pédiatrique, université Paris Denis-Diderot, hôpital Robert-Debré, 48, boulevard Serurier, 75935 Paris cedex 19, France.
| |
Collapse
|
97
|
Arpinati M, Tolomelli G, Bochicchio MT, Castagnetti F, Amabile M, Bandini G, Bonifazi F, Stanzani M, Rosti G, Martinelli G, Baccarani M. Molecular monitoring of BCR-ABL transcripts after allogeneic stem cell transplantation for chronic myeloid leukemia. Biol Blood Marrow Transplant 2013; 19:735-40. [PMID: 23333776 DOI: 10.1016/j.bbmt.2013.01.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Accepted: 01/11/2013] [Indexed: 01/13/2023]
Abstract
The monitoring of minimal residual disease (MRD) through low sensitivity real-time (RT) polymerase chain reaction (PCR) analysis of BCR-ABL transcripts allows early detection of chronic myeloid leukemia (CML) relapse after allogeneic hematopoietic stem cell transplantation (HSCT). The introduction of more sensitive techniques, such as RT quantitative (Q)-PCR, may lead to an overestimation of the risk of CML relapse. In this study, we reviewed the results of peripheral blood RT Q-PCR in CML patients who underwent allogeneic HSCT from 1983 to 2007. In our laboratory, RT Q-PCR analysis was routinely performed since 2002. Eighty-seven of 189 patients had available RT Q-PCR data; 63 patients had at least 3 RT Q-PCR analyses assessable. Fifty-two of 63 patients (83%) had, at least once, detectable transcript levels, but with an BCR-ABL/ABL ratio <.1% defined as <MR3 (molecular remission <0,1%), whereas 11 (17%) had persistent undetectable BCR-ABL transcripts. Six of 52 patients with <MR3 relapsed, defined as BCR-ABL transcript numbers >.1% confirmed by the finding of Ph+ cells in bone marrow. No patients with persistent undetectable transcripts relapsed (P = .19). Relapse did not correlate with the number of occurrences of <MR3 or with the time to the first <MR3 result. Finally, of 46 patients with detectable transcripts who did not relapse, 35 had undetectable transcripts at last contact. RT Q-PCR analysis had low specificity (19%) and low positive predictive value (12%) in predicting relapse of CML patients after allogeneic HSCT. Our data suggest that detection of low BCR-ABL transcript levels by RT Q-PCR analysis has a poor accuracy in defining the risk of CML relapse and should not be considered as the unique indication to treatment. Fluctuation of BCR-ABL transcripts levels is common as late as ≥10 years posttransplantation, possibly suggesting the long-term persistence of CML stem cells.
Collapse
Affiliation(s)
- Mario Arpinati
- Department of Hematology and Oncological Sciences Seragnoli, University of Bologna, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
The role of allogeneic haematopoietic progenitor cell transplantation in patients with diffuse large B-cell non-Hodgkin lymphomas (DLBCL). Bone Marrow Transplant 2013; 48:1271-8. [PMID: 23318539 DOI: 10.1038/bmt.2012.266] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Revised: 11/04/2012] [Accepted: 11/13/2012] [Indexed: 11/08/2022]
Abstract
Despite the undoubted improvement in the prognosis of patients with diffuse large B-cell lymphomas (DLBCLs) with the addition of rituximab in the front-line treatment, a significant proportion of patients still relapse. Salvage immune-chemotherapy followed by high-dose therapy with autologous haematopoietic cell transplantation (auto-HCT) remains the treatment of choice for such patients, especially in those who demonstrate chemosensitive disease. In recent years, allogeneic haematopoietic cell transplantation (allo-HCT) has increasingly been used for patients who are resistant to salvage treatment or relapse after an auto-HCT. Strategies using reduced intensity conditioning regimens have allowed application of this approach to a broader range of patients. PFS is up to 55% with a risk of relapse up to 80% depending on different studies. In multivariate analysis, several factors have been associated with favourable outcome including chemosensitivity of the disease, younger age and Karnofsky performance status at the time of the transplant being the strongest ones. DLIs have shown to induce durable responses in relapsed or progressed disease; however, its role remains controversial as the results are inferior to the responses seen in other haematological malignancies. More recently, the addition of MoAbs in the non-myeloablative conditioning regimens has shown encouraging results. In conclusion, allo-HCT is a feasible option in selective patients with chemosensitive DBCL, as it reduces the risk of relapse; however, this is achieved at the cost of significant non-relapse mortality.
Collapse
|
99
|
Chang YJ, Huang XJ. Donor lymphocyte infusions for relapse after allogeneic transplantation: when, if and for whom? Blood Rev 2012; 27:55-62. [PMID: 23261066 DOI: 10.1016/j.blre.2012.11.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Donor lymphocyte infusion (DLI) using unstimulated leukapheresis is one of the most effective treatment strategies for patients with hematological malignancies; its graft-versus-leukemia effects make it especially effective in chronic myeloid leukemia patients who relapsed after allogeneic stem cell transplantation (allo-HSCT). However, DLI application is limited by the development of graft-versus-host disease and aplasia, and thus cannot be routinely applied for prophylaxis. Therefore, important questions remain to be answered, such as when, and whom to DLI? Recent advances enable DLI using allografts of granulocyte colony-stimulating factor-mobilized peripheral blood progenitor cells; allodepleted donor T cells; and infusions of donor-derived, ex vivo-expanded, CD8(+) cytotoxic T lymphocyte, which can decrease relapse and improve transplant outcomes. Preemptive immunotherapy of relapse was also introduced based on the determination of mixed chimerism and minimal residual disease. In this review, we summarize the latest developments in recent strategies that will affect future DLI efficacy - focusing on the disadvantages and advantages of each protocol for the treatment, preemptive therapy, and prophylaxis of relapse.
Collapse
Affiliation(s)
- Ying-Jun Chang
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Xicheng District, Beijing, China
| | | |
Collapse
|
100
|
Abstract
Bispecific antibodies (BiAbs) offer a unique opportunity to redirect immune effector cells to kill cancer cells. BiAbs combine the benefits of different binding specificities of two monoclonal antibodies (mAbs) into a single construct. This unique feature of BiAbs enables approaches that are not possible with single mAbs. Advances in antibody engineering and antigen profiling of malignant cells have led to the development of a number of BiAb formats and their combinations for redirecting effector cells to tumor targets. There have been significant advances in the design and application of BiAbs for intravenous and local injection.The initial barrier of cytokine storm has been partially overcome by more recent constructs that have improved clinical effectiveness without dose-limiting toxicities. Since the recent revival of BiAbs, there has been multiple, ongoing, phase I/II and III trials, and some promising clinical outcomes have been reported in completed clinical studies. This review focuses on arming T cells with BiAbs to create the 'poor man's cytotoxic lymphocyte'.
Collapse
Affiliation(s)
- Lawrence G Lum
- Department of Oncology, Wayne State University and Barbara Ann Karmanos Cancer Center, Detroit, MI, USA
| | | |
Collapse
|