201
|
Kasakovski D, Xu L, Li Y. T cell senescence and CAR-T cell exhaustion in hematological malignancies. J Hematol Oncol 2018; 11:91. [PMID: 29973238 PMCID: PMC6032767 DOI: 10.1186/s13045-018-0629-x] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 06/06/2018] [Indexed: 12/16/2022] Open
Abstract
T cell senescence has been recognized to play an immunosuppressive role in the aging population and cancer patients. Strategies dedicated to preventing or reversing replicative and premature T cell senescence are required to increase the lifespan of human beings and to reduce the morbidity from cancer. In addition, overcoming the T cell terminal differentiation or senescence from lymphoma and leukemia patients is a promising approach to enhance the effectiveness of adoptive cellular immunotherapy (ACT). Chimeric antigen receptor T (CAR-T) cell and T cell receptor-engineered T (TCR-T) cell therapy highly rely on functionally active T cells. However, the mechanisms which drive T cell senescence remain unclear and controversial. In this review, we describe recent progress for restoration of T cell homeostasis from age-related senescence as well as recovery of T cell activation in hematological malignancies.
Collapse
Affiliation(s)
- Dimitri Kasakovski
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632 China
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Ling Xu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632 China
- Department of Hematology, First Affiliated Hospital, School of Medicine, Jinan University, No. 601 West of Huangpu Avenue, Guangzhou, 510632 China
| | - Yangqiu Li
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632 China
- Department of Hematology, First Affiliated Hospital, School of Medicine, Jinan University, No. 601 West of Huangpu Avenue, Guangzhou, 510632 China
| |
Collapse
|
202
|
Castella B, Foglietta M, Riganti C, Massaia M. Vγ9Vδ2 T Cells in the Bone Marrow of Myeloma Patients: A Paradigm of Microenvironment-Induced Immune Suppression. Front Immunol 2018; 9:1492. [PMID: 30013559 PMCID: PMC6036291 DOI: 10.3389/fimmu.2018.01492] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 06/15/2018] [Indexed: 12/17/2022] Open
Abstract
Vγ9Vδ2 T cells are non-conventional T cells with a natural inclination to recognize and kill cancer cells. Malignant B cells, including myeloma cells, are privileged targets of Vγ9Vδ2 T cells in vitro. However, this inclination is often lost in vivo due to multiple mechanisms mediated by tumor cells and local microenvironment. Multiple myeloma (MM) is a paradigm disease in which antitumor immunity is selectively impaired at the tumor site. By interrogating the immune reactivity of bone marrow (BM) Vγ9Vδ2 T cells to phosphoantigens, we have revealed a very early and long-lasting impairment of Vγ9Vδ2 T-cell immune functions which is already detectable in monoclonal gammopathy of undetermined significance (MGUS) and not fully reverted even in clinical remission after autologous stem cell transplantation. Multiple cell subsets [MM cells, myeloid-derived suppressor cells, regulatory T cells, and BM-derived stromal cells (BMSC)] are involved in Vγ9Vδ2 T-cell inhibition via several immune suppressive mechanisms including the redundant expression of multiple immune checkpoints (ICPs). This review will address some aspects related to the dynamics of ICP expression in the BM of MM patients in relationship to the disease status (MGUS, diagnosis, remission, and relapse) and how this multifaceted ICP expression impairs Vγ9Vδ2 T-cell function. We will also provide some suggestions how to rescue Vγ9Vδ2 T cells from the immune suppression operated by ICP and to recover their antimyeloma immune effector functions at the tumor site.
Collapse
Affiliation(s)
- Barbara Castella
- Laboratorio di Immunologia dei Tumori del Sangue (LITS), Centro Interdipartimentale di Ricerca in Biologia Molecolare (CIRBM), Università degli Studi di Torino, Turin, Italy.,SC Ematologia, AO S. Croce e Carle, Cuneo, Italy
| | - Myriam Foglietta
- Laboratorio di Immunologia dei Tumori del Sangue (LITS), Centro Interdipartimentale di Ricerca in Biologia Molecolare (CIRBM), Università degli Studi di Torino, Turin, Italy.,SC Ematologia, AO S. Croce e Carle, Cuneo, Italy
| | - Chiara Riganti
- Dipartimento di Oncologia, Università degli Studi di Torino, Turin, Italy
| | - Massimo Massaia
- Laboratorio di Immunologia dei Tumori del Sangue (LITS), Centro Interdipartimentale di Ricerca in Biologia Molecolare (CIRBM), Università degli Studi di Torino, Turin, Italy.,SC Ematologia, AO S. Croce e Carle, Cuneo, Italy
| |
Collapse
|
203
|
Tan J, Chen S, Huang J, Chen Y, Yang L, Wang C, Zhong J, Lu Y, Wang L, Zhu K, Li Y. Increased exhausted CD8+T cells with programmed death-1, T-cell immunoglobulin and mucin-domain-containing-3 phenotype in patients with multiple myeloma. Asia Pac J Clin Oncol 2018; 14:e266-e274. [PMID: 29943497 DOI: 10.1111/ajco.13033] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 05/29/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Jiaxiong Tan
- Department of Hematology; First Affiliated Hospital; Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education; Jinan University; Guangzhou China
| | - Shaohua Chen
- Department of Hematology; First Affiliated Hospital; Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education; Jinan University; Guangzhou China
| | - Jingying Huang
- Department of Hematology; First Affiliated Hospital; Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education; Jinan University; Guangzhou China
| | - Youchun Chen
- Department of Hematology; First Affiliated Hospital; Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education; Jinan University; Guangzhou China
| | - Lijian Yang
- Department of Hematology; First Affiliated Hospital; Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education; Jinan University; Guangzhou China
| | - Chunli Wang
- Department of Hematology; First Affiliated Hospital; Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education; Jinan University; Guangzhou China
| | - Jun Zhong
- Department of Hematology; First Affiliated Hospital; Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education; Jinan University; Guangzhou China
| | - Yuhong Lu
- Department of Hematology; First Affiliated Hospital; Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education; Jinan University; Guangzhou China
| | - Liang Wang
- Department of Oncology; First Affiliated Hospital; Jinan University; Guangzhou China
| | - Kanger Zhu
- Department of Hematology; First Affiliated Hospital; Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education; Jinan University; Guangzhou China
| | - Yangqiu Li
- Department of Hematology; First Affiliated Hospital; Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education; Jinan University; Guangzhou China
- Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering; Jinan University; Guangzhou China
| |
Collapse
|
204
|
Atanackovic D, Luetkens T. Biomarkers for checkpoint inhibition in hematologic malignancies. Semin Cancer Biol 2018; 52:198-206. [PMID: 29775689 DOI: 10.1016/j.semcancer.2018.05.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 05/14/2018] [Accepted: 05/14/2018] [Indexed: 01/27/2023]
Abstract
In the past few years we have seen remarkable paradigm shifts in the treatment of many solid tumors due to the introduction of inhibitors targeting immune checkpoints such as PD-1/PD-L1 and CTLA-4. Recent results indicate that checkpoint inhibition also represents a very promising approach for certain types of hematologic malignancies. Unfortunately, treatment with checkpoint inhibitors is also associated with substantial toxicities and high costs and only a subset of patients appears to derive clinical benefit from these treatments. This demonstrates the urgent need for biomarkers for the identification of patient populations that are likely to respond to this type of therapy and/or have fewer side effects. Here, we have reviewed available information on the prognostic and predictive value of biomarkers for anti-CTLA-4 and anti-PD-1/PD-L1 as the most commonly used checkpoint inhibitors. There are currently no reliable biomarkers capable of predicting responses to anti-CTLA-4 agents, such as ipilimumab, in hematologic malignancies. Certain polymorphisms in the CTLA-4 gene, however, seem to have an impact on the patients' outcome, especially in the case of chronic lymphocytic leukemia (CLL). There is now sufficient data supporting PD-L1 expression levels in the tumor tissue as an independent prognostic factor in B cell lymphomas such as diffuse large B-cell lymphoma (DLBCL). Overexpression of PD-L1 in the tumor tissue and elevated serum levels of soluble PD-L1 may also represent adverse prognostic factors in certain subtypes of T cell lymphomas. Finally, expression levels of PD-L1 also seem to predict responses to anti-PD-1/PD-L1 approaches in patients with Hodgkin lymphoma. Future studies will have to further delineate the prognostic/predictive role of PD-L1 expression as a biomarker in hematologic malignancies and may be able to identify confounding variables, which will hopefully to some extent be generalizable to other types of anti-tumor immunotherapies.
Collapse
Affiliation(s)
- Djordje Atanackovic
- Multiple Myeloma Program & Cancer Immunology, Division of Hematology and Hematologic Malignancies, University of Utah/Huntsman Cancer Institute, Salt Lake City, UT, United States.
| | - Tim Luetkens
- Multiple Myeloma Program & Cancer Immunology, Division of Hematology and Hematologic Malignancies, University of Utah/Huntsman Cancer Institute, Salt Lake City, UT, United States
| |
Collapse
|
205
|
Pauza CD, Liou ML, Lahusen T, Xiao L, Lapidus RG, Cairo C, Li H. Gamma Delta T Cell Therapy for Cancer: It Is Good to be Local. Front Immunol 2018; 9:1305. [PMID: 29937769 PMCID: PMC6003257 DOI: 10.3389/fimmu.2018.01305] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 05/25/2018] [Indexed: 12/28/2022] Open
Abstract
Human gamma delta T cells have extraordinary properties including the capacity for tumor cell killing. The major gamma delta T cell subset in human beings is designated Vγ9Vδ2 and is activated by intermediates of isoprenoid biosynthesis or aminobisphosphonate inhibitors of farnesyldiphosphate synthase. Activated cells are potent for killing a broad range of tumor cells and demonstrated the capacity for tumor reduction in murine xenotransplant tumor models. Translating these findings to the clinic produced promising initial results but greater potency is needed. Here, we review the literature on gamma delta T cells in cancer therapy with emphasis on the Vγ9Vδ2 T cell subset. Our goal was to examine obstacles preventing effective Vγ9Vδ2 T cell therapy and strategies for overcoming them. We focus on the potential for local activation of Vγ9Vδ2 T cells within the tumor environment to increase potency and achieve objective responses during cancer therapy. The gamma delta T cells and especially the Vγ9Vδ2 T cell subset, have the potential to overcome many problems in cancer therapy especially for tumors with no known treatment, lacking tumor-specific antigens for targeting by antibodies and CAR-T, or unresponsive to immune checkpoint inhibitors. Translation of amazing work from many laboratories studying gamma delta T cells is needed to fulfill the promise of effective and safe cancer immunotherapy.
Collapse
Affiliation(s)
- C David Pauza
- American Gene Technologies International Inc., Rockville, MD, United States
| | - Mei-Ling Liou
- American Gene Technologies International Inc., Rockville, MD, United States
| | - Tyler Lahusen
- American Gene Technologies International Inc., Rockville, MD, United States
| | - Lingzhi Xiao
- American Gene Technologies International Inc., Rockville, MD, United States
| | - Rena G Lapidus
- Department of Medicine, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Cristiana Cairo
- Institute of Human Virology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Haishan Li
- American Gene Technologies International Inc., Rockville, MD, United States
| |
Collapse
|
206
|
Characterization of CD4 + T cell-mediated cytotoxicity in patients with multiple myeloma. Cell Immunol 2018; 327:62-67. [DOI: 10.1016/j.cellimm.2018.02.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 02/11/2018] [Accepted: 02/11/2018] [Indexed: 12/24/2022]
|
207
|
Tremblay-LeMay R, Rastgoo N, Chang H. Modulating PD-L1 expression in multiple myeloma: an alternative strategy to target the PD-1/PD-L1 pathway. J Hematol Oncol 2018; 11:46. [PMID: 29580288 PMCID: PMC5870495 DOI: 10.1186/s13045-018-0589-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 03/11/2018] [Indexed: 02/08/2023] Open
Abstract
Even with recent advances in therapy regimen, multiple myeloma patients commonly develop drug resistance and relapse. The relevance of targeting the PD-1/PD-L1 axis has been demonstrated in pre-clinical models. Monotherapy with PD-1 inhibitors produced disappointing results, but combinations with other drugs used in the treatment of multiple myeloma seemed promising, and clinical trials are ongoing. However, there have recently been concerns about the safety of PD-1 and PD-L1 inhibitors combined with immunomodulators in the treatment of multiple myeloma, and several trials have been suspended. There is therefore a need for alternative combinations of drugs or different approaches to target this pathway. Protein expression of PD-L1 on cancer cells, including in multiple myeloma, has been associated with intrinsic aggressive features independent of immune evasion mechanisms, thereby providing a rationale for the adoption of new strategies directly targeting PD-L1 protein expression. Drugs modulating the transcriptional and post-transcriptional regulation of PD-L1 could represent new therapeutic strategies for the treatment of multiple myeloma, help potentiate the action of other drugs or be combined to PD-1/PD-L1 inhibitors in order to avoid the potentially problematic combination with immunomodulators. This review will focus on the pathophysiology of PD-L1 expression in multiple myeloma and drugs that have been shown to modulate this expression.
Collapse
Affiliation(s)
- Rosemarie Tremblay-LeMay
- Laboratory Hematology/Laboratory Medicine Program, University Health Network, University of Toronto, Toronto, Canada
| | - Nasrin Rastgoo
- Division of Molecular and Cellular Biology, Toronto General Research Institute, Toronto, Canada
| | - Hong Chang
- Laboratory Hematology/Laboratory Medicine Program, University Health Network, University of Toronto, Toronto, Canada. .,Division of Molecular and Cellular Biology, Toronto General Research Institute, Toronto, Canada. .,Department of Talent Highland, First Affiliated Hospital of Xi'an Jiao Tong University, Xian, China. .,Laboratory Hematology, Toronto General Hospital, 200 Elizabeth Street, 11th floor, Toronto, ON, M5G 2C4, Canada.
| |
Collapse
|
208
|
Liu L, Chang YJ, Xu LP, Zhang XH, Wang Y, Liu KY, Huang XJ. T cell exhaustion characterized by compromised MHC class I and II restricted cytotoxic activity associates with acute B lymphoblastic leukemia relapse after allogeneic hematopoietic stem cell transplantation. Clin Immunol 2018; 190:32-40. [PMID: 29477343 DOI: 10.1016/j.clim.2018.02.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 12/28/2017] [Accepted: 02/22/2018] [Indexed: 01/01/2023]
Abstract
Acute B lymphoblastic leukemia (B-ALL) relapse contributes predominantly to the mortality after allogeneic hematopoietic stem cell transplantation (allo-HSCT). However, the mechanism of B-ALL relapse after allo-HSCT remains unknown. The eradication of leukemia after allo-HSCT largely relies on graft-versus-leukemia (GVL) effects mediated by donor T cells. T cell exhaustion, characterized by the increased expression of inhibitory receptors and impaired function, may suppress GVL effects. In this study, we evaluated whether T cell exhaustion was involved in B-ALL relapse after allo-HSCT. The results showed that CD4+ and CD8+ T cells exhibited increased coexpression of PD-1 and Tim-3, and compromised proliferative capacity, cytokine production and cytotoxic potentials in relapsed patients. Additionally, T cells at the tumor site were more easily exhausted than T cells in the peripheral blood. Moreover, the reversal of T cell exhaustion might correlate with effective anti-leukemic responses after reinduction. These results suggested that T cell exhaustion was associated with B-ALL relapse after allo-HSCT as well as its treatment outcome.
Collapse
Affiliation(s)
- Long Liu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 South Street of Xizhimen, Xicheng District, Beijing 100044, China
| | - Ying-Jun Chang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 South Street of Xizhimen, Xicheng District, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, China
| | - Lan-Ping Xu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 South Street of Xizhimen, Xicheng District, Beijing 100044, China
| | - Xiao-Hui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 South Street of Xizhimen, Xicheng District, Beijing 100044, China
| | - Yu Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 South Street of Xizhimen, Xicheng District, Beijing 100044, China
| | - Kai-Yan Liu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 South Street of Xizhimen, Xicheng District, Beijing 100044, China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 South Street of Xizhimen, Xicheng District, Beijing 100044, China; Peking-Tsinghua Center for Life Sciences, Beijing 100871, China; Collaborative Innovation Center of Hematology, Peking University, China.
| |
Collapse
|
209
|
|
210
|
Ghobrial IM, Detappe A, Anderson KC, Steensma DP. The bone-marrow niche in MDS and MGUS: implications for AML and MM. Nat Rev Clin Oncol 2018; 15:219-233. [PMID: 29311715 DOI: 10.1038/nrclinonc.2017.197] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Several haematological malignancies, including multiple myeloma (MM) and acute myeloid leukaemia (AML), have well-defined precursor states that precede the development of overt cancer. MM is almost always preceded by monoclonal gammopathy of undetermined significance (MGUS), and at least a quarter of all patients with myelodysplastic syndromes (MDS) have disease that evolves into AML. In turn, MDS are frequently anteceded by clonal haematopoiesis of indeterminate potential (CHIP). The acquisition of additional genetic and epigenetic alterations over time clearly influences the increasingly unstable and aggressive behaviour of neoplastic haematopoietic clones; however, perturbations in the bone-marrow microenvironment are increasingly recognized to have key roles in initiating and supporting oncogenesis. In this Review, we focus on the concept that the haematopoietic neoplasia-microenvironment relationship is an intimate rapport between two partners, provide an overview of the evidence supporting a role for the bone-marrow niche in promoting neoplasia, and discuss the potential for niche-specific therapeutic targets.
Collapse
Affiliation(s)
- Irene M Ghobrial
- Division of Hematological Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02115, USA
| | - Alexandre Detappe
- Division of Hematological Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02115, USA
| | - Kenneth C Anderson
- Division of Hematological Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02115, USA
| | - David P Steensma
- Division of Hematological Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02115, USA
| |
Collapse
|
211
|
Brummelman J, Pilipow K, Lugli E. The Single-Cell Phenotypic Identity of Human CD8+ and CD4+ T Cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 341:63-124. [DOI: 10.1016/bs.ircmb.2018.05.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
212
|
Oncolytic virotherapy as an immunotherapeutic strategy for multiple myeloma. Blood Cancer J 2017; 7:640. [PMID: 29208938 PMCID: PMC5802552 DOI: 10.1038/s41408-017-0020-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 10/03/2017] [Accepted: 10/12/2017] [Indexed: 12/22/2022] Open
Abstract
Multiple Myeloma (MM), a clonal malignancy of antibody-producing plasma cells, is the second most common hematologic malignancy and results in significant patient morbidity and mortality. The high degree of immune dysregulation in MM, including T cell imbalances and up-regulation of immunosuppressive checkpoint proteins and myeloid derived suppressor cells, allows this malignancy to escape from host immune control. Despite advances in the therapeutic landscape of MM over the last decade, including the introduction of immunomodulatory drugs, the prognosis for this disease is poor, with less than 50% of patients surviving 5 years. Thus, novel treatment strategies are required. Oncolytic viruses (OV) are a promising new class of therapeutics that rely on tumour specific oncolysis and the generation of a potent adaptive anti-tumour immune response for efficacy. To date, a number of OV have shown efficacy in pre-clinical studies of MM with three reaching early phase clinical trials. OVs represent a rational therapeutic strategy for MM based on (1) their tumour tropism, (2) their ability to potentiate anti-tumour immunity and (3) their ability to be rationally combined with other immunotherapeutic agents to achieve a more robust clinical response.
Collapse
|
213
|
Yao D, Xu L, Tan J, Zhang Y, Lu S, Li M, Lu S, Yang L, Chen S, Chen J, Lai J, Lu Y, Wu X, Zha X, Li Y. Re-balance of memory T cell subsets in peripheral blood from patients with CML after TKI treatment. Oncotarget 2017; 8:81852-81859. [PMID: 29137227 PMCID: PMC5669853 DOI: 10.18632/oncotarget.20965] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 08/27/2017] [Indexed: 01/13/2023] Open
Abstract
T cell immune surveillance is considered an important host protection process for inhibiting carcinogenesis. The full capacity of T cell immune surveillance is dependent on T cell homeostasis, particularly for central memory T (TCM) cells and stem cell memory T (TSCM) cells. In this study, distribution of T cell subsets in peripheral blood from 12 patients with chronic myeloid leukemia (CML) and 12 cases with CML in complete remission (CR) was analyzed using a multicolor flow cytometer, and 16 samples from healthy individuals (HIs) served as control. The proportion of CD8+ TSCM and CD4+ and CD8+ TCM cells were lower, while CD4+ effector memory T (TEM) cells and CD4+ and CD8+ terminal effector T (TEF) cells were higher in CML patients compared with HIs. Moreover, the proportion of CD8+CD28- T cells, which were found to have the immune suppressive function, increased in the naive T (TN) cell and TCM subsets in CML patients compared with HIs. Our study reveals that elimination of leukemia cells by treating with tyrosine kinase inhibitors (TKIs) restores the memory T cell distribution from a skewed pattern in CML patients who are under leukemia burden, indicating that leukemia-specific immune responses mediated by T cells might be induced and maintained in CML patients, however, these responsive T cells might gradually become exhausted due to the continued existence of leukemia cells and their environment; therefore, T cell activation using a different approach remains a key point for enhancing global T cell immunity in CML patients, even for those with CR status.
Collapse
Affiliation(s)
- Danlin Yao
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, School of Medicine, Jinan University, Guangzhou, China.,Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Ling Xu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, School of Medicine, Jinan University, Guangzhou, China.,Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Jiaxiong Tan
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, School of Medicine, Jinan University, Guangzhou, China.,Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Yikai Zhang
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, School of Medicine, Jinan University, Guangzhou, China.,Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Shuai Lu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, School of Medicine, Jinan University, Guangzhou, China
| | - Mingde Li
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, School of Medicine, Jinan University, Guangzhou, China.,Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Sichun Lu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, School of Medicine, Jinan University, Guangzhou, China.,Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Lijian Yang
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, School of Medicine, Jinan University, Guangzhou, China
| | - Shaohua Chen
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, School of Medicine, Jinan University, Guangzhou, China
| | - Jie Chen
- Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Jing Lai
- Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Yuhong Lu
- Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Xiuli Wu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, School of Medicine, Jinan University, Guangzhou, China.,Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Xianfeng Zha
- Department of Clinical Laboratory, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Yangqiu Li
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, School of Medicine, Jinan University, Guangzhou, China.,Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, China
| |
Collapse
|
214
|
Gay F, D'Agostino M, Giaccone L, Genuardi M, Festuccia M, Boccadoro M, Bruno B. Immuno-oncologic Approaches: CAR-T Cells and Checkpoint Inhibitors. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2017; 17:471-478. [DOI: 10.1016/j.clml.2017.06.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 06/08/2017] [Indexed: 01/21/2023]
|
215
|
Franssen LE, Roeven MWH, Hobo W, Doorn R, Oostvogels R, Falkenburg JHF, van de Donk NW, Kester MGD, Fredrix H, Westinga K, Slaper-Cortenbach I, Spierings E, Kersten MJ, Dolstra H, Mutis T, Schaap N, Lokhorst HM. A phase I/II minor histocompatibility antigen-loaded dendritic cell vaccination trial to safely improve the efficacy of donor lymphocyte infusions in myeloma. Bone Marrow Transplant 2017; 52:1378-1383. [PMID: 28581468 DOI: 10.1038/bmt.2017.118] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 04/15/2017] [Accepted: 04/20/2017] [Indexed: 12/22/2022]
Abstract
Allogeneic stem cell transplantation (allo-SCT) with or without donor lymphocyte infusions (DLI) is the only curative option for several hematological malignancies. Unfortunately, allo-SCT is often associated with GvHD, and patients often relapse. We therefore aim to improve the graft-versus-tumor effect, without increasing the risk of GvHD, by targeting hematopoietic lineage-restricted and tumor-associated minor histocompatibility antigens using peptide-loaded dendritic cell (DC) vaccinations. In the present multicenter study, we report the feasibility, safety and efficacy of this concept. We treated nine multiple myeloma patients with persistent or relapsed disease after allo-SCT and a previous DLI, with donor monocyte-derived mHag-peptide-loaded DC vaccinations combined with a second DLI. Vaccinations were well tolerated and no occurrence of GvHD was observed. In five out of nine patients, we were able to show the induction of mHag-specific CD8+ T cells in peripheral blood. Five out of nine patients, of which four developed mHag-specific T cells, showed stable disease (SD) for 3.5-10 months. This study shows that mHag-based donor monocyte-derived DC vaccination combined with DLI is safe, feasible and capable of inducing objective mHag-specific T-cell responses. Future research should focus on further improvement of the vaccination strategy, toward translating the observed T-cell responses into robust clinical responses.
Collapse
Affiliation(s)
- L E Franssen
- Department of Hematology, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - M W H Roeven
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - W Hobo
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - R Doorn
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - R Oostvogels
- Department of Hematology, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - J H F Falkenburg
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - N W van de Donk
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - M G D Kester
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - H Fredrix
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - K Westinga
- Cell Therapy Facility, University Medical Center Utrecht, Utrecht, The Netherlands
| | - I Slaper-Cortenbach
- Cell Therapy Facility, University Medical Center Utrecht, Utrecht, The Netherlands
| | - E Spierings
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - M J Kersten
- Department of Hematology, Amsterdam Medical Center, Amsterdam, The Netherlands
| | - H Dolstra
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - T Mutis
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - N Schaap
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - H M Lokhorst
- Department of Hematology, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
216
|
Shallis RM, Terry CM, Lim SH. The multi-faceted potential of CD38 antibody targeting in multiple myeloma. Cancer Immunol Immunother 2017; 66:697-703. [PMID: 28341874 PMCID: PMC11029060 DOI: 10.1007/s00262-017-1990-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Accepted: 03/16/2017] [Indexed: 10/19/2022]
Abstract
CD38, an adenine dinucleotide phosphate (ADP) ribose cyclase and a cyclic ADP ribose hydrolase, is widely expressed on the surface of multiple myeloma (MM) cells. It is known to play a pivotal role in the downstream pathways that mediate MM cell growth, signal transduction, and adhesion. The clinical use of CD38 monoclonal antibodies (MoAbs), such as daratumumab, either as monotherapy or in combination with other anti-MM agents, has produced impressive results in patients who have failed standard MM therapy. CD38 MoAbs exhibit several cytotoxic mechanisms on MM cells. In addition to the classical effector mechanisms associated with antibody therapy, CD38 MoAbs induce MM apoptosis and clonal T-cell expansion. Here, we summarize the results of some pivotal clinical studies using a human CD38 MoAb, daratumumab, in patients with MM, discuss the anti-MM effector mechanisms induced by CD38 MoAbs, and review the potential tumor antigens that may be suitable targets for immunotherapy of MM. Finally, we present a paradigm of immunotherapy for MM patients using CD38 MoAbs followed by GM-CSF and an immune checkpoint inhibitor in patients who have undergone high dose chemotherapy and autologous stem cell transplant. CD38 MoAbs have emerged as a novel and ultimately very promising immunotherapeutic agent for MM because of its ability to induce MM cytotoxicity through both arms of the adaptive immune responses.
Collapse
Affiliation(s)
- Rory M Shallis
- Division of Hematology and Oncology, Rhode Island Hospital/Brown University Warren Alpert Medical School, Room 140, APC Building, 593 Eddy Street, Providence, RI, 02903, USA
| | - Christopher M Terry
- Division of Hematology and Oncology, Rhode Island Hospital/Brown University Warren Alpert Medical School, Room 140, APC Building, 593 Eddy Street, Providence, RI, 02903, USA
| | - Seah H Lim
- Division of Hematology and Oncology, Rhode Island Hospital/Brown University Warren Alpert Medical School, Room 140, APC Building, 593 Eddy Street, Providence, RI, 02903, USA.
| |
Collapse
|