1
|
Sayadmanesh A, Azadbakht M, Yari K, Abedelahi A, Shafaei H, Shanehbandi D, Baradaran B, Basiri M. Characterization of CAR T Cells Manufactured Using Genetically Engineered Artificial Antigen Presenting Cells. CELL JOURNAL 2023; 25:674-687. [PMID: 37865876 PMCID: PMC10591261 DOI: 10.22074/cellj.2023.2001712.1304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/22/2023] [Accepted: 07/01/2023] [Indexed: 10/23/2023]
Abstract
OBJECTIVE Chimeric antigen receptor (CAR) T cell therapy has recently emerged as a promising approach for the treatment of different types of cancer. Improving CAR T cell manufacturing in terms of costs and product quality is an important concern for expanding the accessibility of this therapy. One proposed strategy for improving T cell expansion is to use genetically engineered artificial antigen presenting cells (aAPC) expressing a membrane-bound anti-CD3 for T cell activation. The aim of this study was to characterize CAR T cells generated using this aAPC-mediated approach in terms of expansion efficiency, immunophenotype, and cytotoxicity. MATERIALS AND METHODS In this experimental study, we generated an aAPC line by engineering K562 cells to express a membrane-bound anti-CD3 (mOKT3). T cell activation was performed by co-culturing PBMCs with either mitomycin C-treated aAPCs or surface-immobilized anti-CD3 and anti-CD28 antibodies. Untransduced and CD19-CARtransduced T cells were characterized in terms of expansion, activation markers, interferon gamma (IFN-γ) secretion, CD4/CD8 ratio, memory phenotype, and exhaustion markers. Cytotoxicity of CD19-CAR T cells generated by aAPCs and antibodies were also investigated using a bioluminescence-based co-culture assay. RESULTS Our findings showed that the engineered aAPC line has the potential to expand CAR T cells similar to that using the antibody-based method. Although activation with aAPCs leads to a higher ratio of CD8+ and effector memory T cells in the final product, we did not observe a significant difference in IFN-γ secretion, cytotoxic activity or exhaustion between CAR T cells generated with aAPC or antibodies. CONCLUSION Our results show that despite the differences in the immunophenotypes of aAPC and antibody-based CAR T cells, both methods can be used to manufacture potent CAR T cells. These findings are instrumental for the improvement of the CAR T cell manufacturing process and future applications of aAPC-mediated expansion of CAR T cells.
Collapse
Affiliation(s)
- Ali Sayadmanesh
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohamad Azadbakht
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| | - Kheirollah Yari
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ali Abedelahi
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hajar Shafaei
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohsen Basiri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
- Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
2
|
Bastin DJ, Quizi J, Kennedy MA, Kekre N, Auer RC. Current challenges in the manufacture of clinical-grade autologous whole cell vaccines for hematological malignancies. Cytotherapy 2022; 24:979-989. [PMID: 35562303 DOI: 10.1016/j.jcyt.2022.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/21/2022] [Accepted: 03/21/2022] [Indexed: 11/03/2022]
Abstract
Autologous whole cell vaccines use a patient's own tumor cells as a source of antigen to elicit an anti-tumor immune response in vivo. Recently, the authors conducted a systematic review of clinical trials employing these products in hematological cancers that showed a favorable safety profile and trend toward efficacy. However, it was noted that manufacturing challenges limit both the efficacy and clinical implementation of these vaccine products. In the current literature review, the authors sought to define the issues surrounding the manufacture of autologous whole cell products for hematological cancers. The authors describe key factors, including the acquisition, culture, cryopreservation and transduction of malignant cells, that require optimization for further advancement of the field. Furthermore, the authors provide a summary of pre-clinical work that informs how the identified challenges may be overcome. The authors also highlight areas in which future basic research would be of benefit to the field. The goal of this review is to provide a roadmap for investigators seeking to advance the field of autologous cell vaccines as it applies to hematological malignancies.
Collapse
Affiliation(s)
- Donald J Bastin
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada; Schulich School of Medicine, Western University, London, Canada
| | - Jennifer Quizi
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Michael A Kennedy
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Natasha Kekre
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada; Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Rebecca C Auer
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada; Faculty of Medicine, University of Ottawa, Ottawa, Canada; Department of Surgery, University of Ottawa, Ottawa, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada.
| |
Collapse
|
3
|
Viswanath DI, Liu HC, Capuani S, Vander Pol RS, Saunders SZ, Chua CYX, Grattoni A. Engineered implantable vaccine platform for continuous antigen-specific immunomodulation. Biomaterials 2022; 281:121374. [PMID: 35066287 PMCID: PMC8865051 DOI: 10.1016/j.biomaterials.2022.121374] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/03/2022] [Accepted: 01/12/2022] [Indexed: 02/03/2023]
Abstract
Cancer vaccines harness the host immune system to generate antigen-specific antitumor immunity for long-term tumor elimination with durable immunomodulation. Commonly investigated strategies reintroduce ex vivo autologous dendritic cells (DCs) but have limited clinical adoption due to difficulty in manufacturing, delivery and low clinical efficacy. To combat this, we designed the "NanoLymph", an implantable subcutaneous device for antigen-specific antitumor immunomodulation. The NanoLymph consists of a dual-reservoir platform for sustained release of immune stimulants via a nanoporous membrane and hydrogel-encapsulated antigens for local immune cell recruitment and activation, respectively. Here, we present the development and characterization of the NanoLymph as well as efficacy validation for immunomodulation in an immunocompetent murine model. Specifically, we established the NanoLymph biocompatibility and mechanical stability. Further, we demonstrated minimally invasive transcutaneous refilling of the drug reservoir in vivo for prolonging drug release duration. Importantly, our study demonstrated that local elution of two drugs (GMCSF and Resiquimod) generates an immune stimulatory microenvironment capable of local DC recruitment and activation and generation of antigen-specific T lymphocytes within 14 days. In summary, the NanoLymph approach can achieve in situ immunomodulation, presenting a viable strategy for therapeutic cancer vaccines.
Collapse
Affiliation(s)
- Dixita Ishani Viswanath
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA; Texas A&M University College of Medicine, Bryan & Houston, TX, USA
| | - Hsuan-Chen Liu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Simone Capuani
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA; University of Chinese Academy of Science (UCAS), Shijingshan, 19 Yuquan Road, Beijing, 100049, China
| | | | | | | | - Alessandro Grattoni
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA; Department of Surgery, Houston Methodist Hospital, Houston, TX, USA; Department of Radiation Oncology, Houston Methodist Hospital, Houston, TX, USA.
| |
Collapse
|
4
|
Viswanath DI, Liu HC, Huston DP, Chua CYX, Grattoni A. Emerging biomaterial-based strategies for personalized therapeutic in situ cancer vaccines. Biomaterials 2022; 280:121297. [PMID: 34902729 PMCID: PMC8725170 DOI: 10.1016/j.biomaterials.2021.121297] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 11/19/2021] [Accepted: 11/29/2021] [Indexed: 01/03/2023]
Abstract
Landmark successes in oncoimmunology have led to development of therapeutics boosting the host immune system to eradicate local and distant tumors with impactful tumor reduction in a subset of patients. However, current immunotherapy modalities often demonstrate limited success when involving immunologically cold tumors and solid tumors. Here, we describe the role of various biomaterials to formulate cancer vaccines as a form of cancer immunotherapy, seeking to utilize the host immune system to activate and expand tumor-specific T cells. Biomaterial-based cancer vaccines enhance the cancer-immunity cycle by harnessing cellular recruitment and activation against tumor-specific antigens. In this review, we discuss biomaterial-based vaccine strategies to induce lymphocytic responses necessary to mediate anti-tumor immunity. We focus on strategies that selectively attract dendritic cells via immunostimulatory gradients, activate them against presented tumor-specific antigens, and induce effective cross-presentation to T cells in secondary lymphoid organs, thereby generating immunity. We posit that personalized cancer vaccines are promising targets to generate long-term systemic immunity against patient- and tumor-specific antigens to ensure long-term cancer remission.
Collapse
Affiliation(s)
- Dixita Ishani Viswanath
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA; Texas A&M University College of Medicine, Bryan & Houston, TX, USA
| | - Hsuan-Chen Liu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - David P Huston
- Texas A&M University College of Medicine, Bryan & Houston, TX, USA
| | | | - Alessandro Grattoni
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA; Department of Surgery, Houston Methodist Hospital, Houston, TX, USA; Department of Radiation Oncology, Houston Methodist Hospital, Houston, TX, USA.
| |
Collapse
|
5
|
Bastin DJ, Khan ST, Montroy J, Kennedy MA, Forbes N, Martel AB, Baker L, Gresham L, Boucher DM, Wong B, Shorr R, Diallo JS, Fergusson DA, Lalu MM, Auer RC, Kekre N. Safety and efficacy of autologous whole cell vaccines in hematologic malignancies: A systematic review and meta-analysis. Hematol Oncol 2021; 39:448-464. [PMID: 33963789 DOI: 10.1002/hon.2875] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 04/26/2021] [Indexed: 01/07/2023]
Abstract
Autologous cell vaccines use a patient's tumor cells to stimulate a broad antitumor response in vivo. This approach shows promise for treating hematologic cancers in early phase clinical trials, but overall safety and efficacy remain poorly described. We conducted a systematic review assessing the use of autologous cell vaccination in treating hematologic cancers. Primary outcomes of interest were safety and clinical response, with secondary outcomes including survival, relapse rate, correlative immune assays and health-quality related metrics. We performed a search of MEDLINE, Embase and the Cochrane Register of Controlled Trials including any interventional trial employing an autologous, whole cell product in any hematologic malignancy. Risk of bias was assessed using a modified Institute of Health Economics tool. Across 20 single arm studies, only 341 of 592 enrolled participants received one or more vaccinations. Primary reasons for not receiving vaccination included rapid disease progression/death and manufacturing challenges. Overall, few high-grade adverse events were observed. One death was reported and attributed to a GM-CSF producing allogeneic cell line co-administered with the autologous vaccine. Of 58 evaluable patients, the complete response rate was 21.0% [95% CI, 10.4%-37.8%)] and overall response rate was 35.8% (95% CI, 24.4%-49.0%). Of 97 evaluable patients for survival, the 5-years overall survival rate was 64.9% (95% CI, 52.6%-77.2%) and disease-free survival was 59.7% (95% CI, 47.7%-71.7%). We conclude that, in hematologic malignancies, based on limited available data, autologous cell vaccines are safe and display a trend towards efficacy but that challenges exist in vaccine manufacture and administration.
Collapse
Affiliation(s)
- Donald J Bastin
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Schulich School of Medicine, Western University, London, ON, Canada
| | - Sarwat T Khan
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Joshua Montroy
- Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Michael A Kennedy
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Nicole Forbes
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Andre B Martel
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Surgery, University of Ottawa, Ottawa, Ontario, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Laura Baker
- Department of Surgery, University of Ottawa, Ottawa, Ontario, Canada
| | - Louise Gresham
- Department of Surgery, University of Ottawa, Ottawa, Ontario, Canada
| | - Dominique M Boucher
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Boaz Wong
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Risa Shorr
- Learning Services, The Ottawa Hospital, Ottawa, ON, Canada
| | - Jean-Simon Diallo
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Dean A Fergusson
- Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada.,School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| | - Manoj M Lalu
- Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada.,Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada.,Regenerative Medicine Program, The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Rebecca C Auer
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Surgery, University of Ottawa, Ottawa, Ontario, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Natasha Kekre
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada.,Department of Medicine and The Ottawa Hospital, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
6
|
Wu M, Wang S, Chen JY, Zhou LJ, Guo ZW, Li YH. Therapeutic cancer vaccine therapy for acute myeloid leukemia. Immunotherapy 2021; 13:863-877. [PMID: 33955237 DOI: 10.2217/imt-2020-0277] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Antitumor function of the immune system has been harnessed to eradicate tumor cells as cancer therapy. Therapeutic cancer vaccines aim to help immune cells recognize tumor cells, which are difficult to target owing to immune escape. Many attempts at vaccine designs have been conducted throughout the last decades. In addition, as the advanced understanding of immunosuppressive mechanisms mediated by tumor cells, combining cancer vaccines with other immune therapies seems to be more efficient for cancer treatment. Acute myeloid leukemia (AML) is the most common acute leukemia in adults with poor prognosis. Evidence has shown T-cell-mediated immune responses in AML, which encourages the utility of immune therapies in AML. This review discusses cancer vaccines in AML from vaccine design as well as recent progress in vaccination combination with other immune therapies.
Collapse
Affiliation(s)
- Ming Wu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.,Department of Hematology, Zhongshan People's Hospital, Zhongshan 528400, China
| | - Sheng Wang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Jian-Yu Chen
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Li-Juan Zhou
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Zi-Wen Guo
- Department of Hematology, Zhongshan People's Hospital, Zhongshan 528400, China
| | - Yu-Hua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| |
Collapse
|
7
|
Xie X, Hu Y, Ye T, Chen Y, Zhou L, Li F, Xi X, Wang S, He Y, Gao X, Wei W, Ma G, Li Y. Therapeutic vaccination against leukaemia via the sustained release of co-encapsulated anti-PD-1 and a leukaemia-associated antigen. Nat Biomed Eng 2021; 5:414-428. [PMID: 33046865 DOI: 10.1038/s41551-020-00624-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 09/03/2020] [Indexed: 02/08/2023]
Abstract
Therapeutic leukaemia vaccines have shown modest potency. Here, we show that the co-encapsulation of a leukaemia-associated epitope peptide highly expressed in leukaemia patients and of the immune checkpoint inhibitor anti-programmed-cell-death-protein-1 (anti-PD-1) in degradable poly(lactic acid) microcapsules resulted in the sustained release of the peptide and of the antibody, which led to the recruitment of activated antigen-presenting cells to the injection site, their uptake of the peptide and the transportation of the anti-PD-1 antibody to lymph nodes, enhancing the expansion of epitope-specific T cells and the activation of cytotoxic T cells. After single subcutaneous injections of vaccine formulations with different epitope peptides, mice bearing leukaemia xenografts derived from humanized cell lines or from primary cells from patients showed better therapeutic outcomes than mice receiving repeated injections of free antigen, antibody and a commercial adjuvant. The sustained release of a tumour-associated peptide and of anti-PD-1 may represent a generalizable strategy for boosting antitumour immune responses to leukaemia.
Collapse
Affiliation(s)
- Xiaoling Xie
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, P R China.,State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P R China
| | - Yuxing Hu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, P R China
| | - Tong Ye
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P R China.,University of Chinese Academy of Sciences, Beijing, P R China
| | - Yiran Chen
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, P R China
| | - Lijuan Zhou
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, P R China
| | - Feng Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P R China.,University of Chinese Academy of Sciences, Beijing, P R China
| | - Xiaobo Xi
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P R China.,University of Chinese Academy of Sciences, Beijing, P R China
| | - Shuang Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P R China
| | - Yanjie He
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, P R China
| | - Xiaoyong Gao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P R China
| | - Wei Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P R China. .,University of Chinese Academy of Sciences, Beijing, P R China.
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P R China. .,University of Chinese Academy of Sciences, Beijing, P R China.
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, P R China. .,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, P R China.
| |
Collapse
|
8
|
Schmidts A, Marsh LC, Srivastava AA, Bouffard AA, Boroughs AC, Scarfò I, Larson RC, Bedoya F, Choi BD, Frigault MJ, Bailey SR, Leick MB, Vatsa S, Kann MC, Prew MS, Kleinstiver BP, Joung JK, Maus MV. Cell-based artificial APC resistant to lentiviral transduction for efficient generation of CAR-T cells from various cell sources. J Immunother Cancer 2020; 8:jitc-2020-000990. [PMID: 32900862 PMCID: PMC7477986 DOI: 10.1136/jitc-2020-000990] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/02/2020] [Indexed: 01/14/2023] Open
Abstract
Background Adoptive cell therapy with chimeric antigen receptor T cells (CAR-T) has become a standard treatment for patients with certain aggressive B cell malignancies and holds promise to improve the care of patients suffering from numerous other cancers in the future. However, the high manufacturing cost of CAR-T cell therapies poses a major barrier to their broader clinical application. Among the key cost drivers of CAR-T production are single-use reagents for T cell activation and clinical-grade viral vector. The presence of variable amounts of contaminating monocytes in the starting material poses an additional challenge to CAR-T manufacturing, since they can impede T cell stimulation and transduction, resulting in manufacturing failure. Methods We created K562-based artificial antigen-presenting cells (aAPC) with genetically encoded T cell stimulation and costimulation that represent an inexhaustible source for T cell activation. We additionally disrupted endogenous expression of the low-density lipoprotein receptor (LDLR) on these aAPC (aAPC-ΔLDLR) using CRISPR-Cas9 gene editing nucleases to prevent inadvertent lentiviral transduction and avoid the sink effect on viral vector during transduction. Using various T cell sources, we produced CD19-directed CAR-T cells via aAPC-ΔLDLR-based activation and tested their in vitro and in vivo antitumor potency against B cell malignancies. Results We found that lack of LDLR expression on our aAPC-ΔLDLR conferred resistance to lentiviral transduction during CAR-T production. Using aAPC-ΔLDLR, we achieved efficient expansion of CAR-T cells even from unpurified starting material like peripheral blood mononuclear cells or unmanipulated leukapheresis product, containing substantial proportions of monocytes. CD19-directed CAR-T cells that we produced via aAPC-ΔLDLR-based expansion demonstrated potent antitumor responses in preclinical models of acute lymphoblastic leukemia and B-cell lymphoma. Conclusions Our aAPC-ΔLDLR represent an attractive approach for manufacturing of lentivirally transduced T cells that may be simpler and more cost efficient than currently available methods.
Collapse
Affiliation(s)
- Andrea Schmidts
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Leah C Marsh
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ambike A Srivastava
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Amanda A Bouffard
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Angela C Boroughs
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Irene Scarfò
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Rebecca C Larson
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Felipe Bedoya
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Bryan D Choi
- Cellular Immunotherapy Program and Department of Neurosurgery, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Matthew J Frigault
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Stefanie R Bailey
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Mark B Leick
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Sonika Vatsa
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Michael C Kann
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Michelle S Prew
- Molecular Pathology Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Benjamin P Kleinstiver
- Center for Genomic Medicine and Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| | - J Keith Joung
- Molecular Pathology Unit, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA .,Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Khan ST, Montroy J, Forbes N, Bastin D, Kennedy MA, Diallo JS, Kekre N, Fergusson DA, Lalu M, Auer RC. Safety and efficacy of autologous tumour cell vaccines as a cancer therapeutic to treat solid tumours and haematological malignancies: a meta-analysis protocol for two systematic reviews. BMJ Open 2020; 10:e034714. [PMID: 32518209 PMCID: PMC7282323 DOI: 10.1136/bmjopen-2019-034714] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Autologous cancer cell vaccines are promising personalised immunotherapeutic options for solid and haematological malignancies that uses the patient's own cells to arm an immune response. Evidence suggests that among patients receiving these vaccines, those who mount an immune response against their own tumour cells have better prognosis, and a myriad of preclinical studies have demonstrated the same. Recently, two autologous cell vaccines Vigil and OncoVAX have made it to phase III clinical trials. Here, we outline a protocol to be used for two separate systematic reviews using a parallel approach for inclusion criteria, data extraction and analysis for autologous cell vaccines in (1) solid and (2) haematological malignancies. We aim to review evidence from controlled and uncontrolled interventional studies of autologous cell vaccines administered to patients with cancer to determine their historical efficacy (with or without associated adjuvants or modifications) with clinical response rates and safety outcomes being of particular importance. METHODS AND ANALYSIS We will search MEDLINE (OVID interface, including In-Process and Epub Ahead of Print), Embase (OVID interface) and the Cochrane Central Register of Controlled Trials (Wiley interface) for articles published from 1947 until 30 July 2018 (date search was performed). Studies will be screened first by title and abstract, then by full-text in duplicate. Interventional trials that report the use of an autologous cell vaccine to patients with cancer of any age will be included. The primary outcomes of interest in this review are clinical response (complete or overall/objective response) and safety outcomes (adverse events). Secondary outcomes include immune response, disease-free survival and overall survival. The risk of bias within studies will be assessed using the appropriate Cochrane Risk of Bias tool. If appropriate, a random effects meta-analysis will be performed to synthesise the data and report summary estimates of effect. Statistical heterogeneity will be assessed using the I2 statistic. ETHICS AND DISSEMINATION Ethics approval is not required for this systematic review protocol as the review will solely use published literature. Results will be submitted to peer-reviewed journals for publication and presented to relevant stakeholders and scientific meetings. PROSPERO REGISTRATION NUMBER CRD42019140187.
Collapse
Affiliation(s)
- Sarwat T Khan
- Cancer Therapeutic Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Joshua Montroy
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Nicole Forbes
- Cancer Therapeutic Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Donald Bastin
- Cancer Therapeutic Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Michael A Kennedy
- Cancer Therapeutic Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Jean-Simon Diallo
- Cancer Therapeutic Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Natasha Kekre
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Blood and Marrow Transplant Program, Ottawa Hospital General Campus, Ottawa, Ontario, Canada
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Dean A Fergusson
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Manoj Lalu
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Anaesthesiology and Pain Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Rebecca C Auer
- Cancer Therapeutic Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Department of Surgery, Ottawa Hospital General Campus, Ottawa, Ontario, Canada
| |
Collapse
|
10
|
Ji YS, Park SK, Ryu S. Whole leukemia cell vaccines: Past progress and future directions. Vaccine 2020; 38:3811-3820. [PMID: 32280046 DOI: 10.1016/j.vaccine.2020.03.042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 03/17/2020] [Accepted: 03/21/2020] [Indexed: 11/28/2022]
Abstract
It has long been recognized that allogeneic hematopoietic stem cell transplantation can reduce the risk of leukemia relapse by inducing the graft-versus-leukemia effect. However, allogeneic stem cell transplantation is also known to be able to cause graft-versus-host disease, which can cause considerable morbidity and even mortality in patients receiving allogeneic hematopoietic stem cell transplantation. Therefore, to elicit leukemia-specific immune responses without alloimmune reaction, the possibilities of active immunotherapy methods such as leukemia vaccines have been studied for decades. Among various types of leukemia vaccines, whole leukemia cell vaccines are known to be able to induce immune responses against multiple unknown antigens without the need for adoptive transfer of dendritic cells. In this review, we will discuss the past progress of whole leukemia cell vaccines, with a focus on strategies to enhance their immunogenicity. We will also present the future directions of whole leukemia cell vaccines along with addressing newly emerging concepts, such as immunogenic cell death and necroptosis. We will not discuss in detail other factors that can reduce the therapeutic efficacy of whole leukemia cell vaccines such as various immunosuppressive mechanisms of leukemia.
Collapse
Affiliation(s)
- Young Sok Ji
- Department of Pathology, School of Medicine, Soonchunhyang University, 25 Bongjeong-ro, Dongnam-gu, Cheonan-si, Chungcheongnam-do 31151, Republic of Korea.
| | - Seong Kyu Park
- Division of Hemato-Oncology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, 170 Jomaru-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 14584, Republic of Korea.
| | - Seongho Ryu
- Department of Pathology, School of Medicine, Soonchunhyang University, 25 Bongjeong-ro, Dongnam-gu, Cheonan-si, Chungcheongnam-do 31151, Republic of Korea; Soonchunhyang Institute of Medi-bio Sciences (SIMS), Soonchunhyang University, 25 Bongjeong-ro, Dongnam-gu, Cheonan-si, Chungcheongnam-do 31151, Republic of Korea.
| |
Collapse
|
11
|
IL-15/IL-15Rα/CD80-expressing AML cell vaccines eradicate minimal residual disease in leukemic mice. Blood Adv 2019; 2:3177-3192. [PMID: 30482760 DOI: 10.1182/bloodadvances.2018019026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 10/14/2018] [Indexed: 12/17/2022] Open
Abstract
Engineered autologous acute myeloid leukemia (AML) cells present multiple leukemia-associated and patient-specific antigens and as such hold promise as immunotherapeutic vaccines. However, prior vaccines have not reliably induced effective antileukemic immunity, in part because AML blasts have immune inhibitory effects and lack expression of the critical costimulatory molecule CD80. To enhance induction of leukemia-specific cytolytic activity, 32Dp210 murine AML cells were engineered to express either CD80 alone, or the immunostimulatory cytokine interleukin-15 (IL-15) with its receptor α (IL-15Rα), or heterodimeric IL-15/IL-15Rα together with CD80 and tested as irradiated cell vaccines. IL-15 is a γc-chain cytokine, with unique properties suited to stimulating antitumor immunity, including stimulation of both natural killer and CD8+ memory T cells. Coexpression of IL-15 and IL-15Rα markedly increases IL-15 stability and secretion. Non-tumor-bearing mice vaccinated with irradiated 32Dp210-IL-15/IL-15Rα/CD80 and challenged with 32Dp210 leukemia had greater survival than did mice treated with 32Dp210-CD80 or 32Dp210-IL-15/IL-15Rα vaccines, whereas no unvaccinated mice inoculated with leukemia survived. In mice with established leukemia, treatment with 32Dp210-IL-15/IL-15Rα/CD80 vaccination stimulated unprecedented antileukemic immunity enabling 80% survival, an effect that was abrogated by anti-CD8 antibody-mediated depletion in vivo. Because, clinically, AML vaccines are administered as postremission therapy, we established a novel model in which mice with high leukemic burdens were treated with cytotoxic therapy to induce remission (<5% marrow blasts). Postremission vaccination with 32Dp210-IL-15/IL-15Rα/CD80 achieved 50% overall survival in these mice, whereas all unvaccinated mice achieving remission subsequently relapsed. These studies demonstrate that combined expression of IL-15/IL-15Rα and CD80 by syngeneic AML vaccines stimulates effective and long-lasting antileukemic immunity.
Collapse
|
12
|
Role of Interferon (IFN)α in “Cocktails” for the Generation of (Leukemia-derived) Dendritic Cells (DCleu) From Blasts in Blood From Patients (pts) With Acute Myeloid Leukemia (AML) and the Induction of Antileukemic Reactions. J Immunother 2019; 42:143-161. [DOI: 10.1097/cji.0000000000000266] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
13
|
Pre-immunization of donor lymphocytes with GITR agonistic antibody enhances antitumor immunity in autologous hematopoietic stem cell transplantation. Biochem Biophys Res Commun 2019; 509:96-101. [PMID: 30579597 DOI: 10.1016/j.bbrc.2018.12.069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 12/11/2018] [Indexed: 01/24/2023]
Abstract
The lymphopenic condition following autologous hematopoietic stem cell transplantation (HSCT) enhances the proliferation of T cells by engaging tumor-associated antigens, leading to the alteration of the T-cell repertoire towards antitumor immunity. However, cure by autologous HSCT alone have rarely occurred in the clinical setting. Since tumor-reactive lymphocytes preferentially proliferate during reconstitution of the immune system, we examined whether the priming of donor lymphocytes can strengthen the antitumor effect by HSCT in a CT26 murine colon cancer model. The systemic administration of an anti-glucocorticoid-induced TNF receptor (GITR) agonistic antibody (Ab) significantly increased the number of CT26-responsive T cells but not that of auto-reactive lymphocytes in donor mice. The infusion of non-primed and GITR Ab-primed donor lymphocytes suppressed the CT26 tumor growth, and only the primed lymphocytes eliminated tumors in all the treated mice. The frequency of CT26-responsive T cells was elevated in recipient mice infused with both primed and non-primed lymphocytes until 4 weeks after transplantation, while the frequency in recipients with primed lymphocytes was markedly elevated compared with that in mice harboring non-primed lymphocytes at 2 weeks. The frequencies of regulatory T cells and myeloid-derived suppressor cells were elevated in recipient mice infused with primed and non-primed lymphocytes 2 weeks after transplantation, and returned to normal levels by week 4. The combination of autologous HSCT with pre-immunization of donor lymphocytes is a promising strategy to induce strong antitumor immunity.
Collapse
|
14
|
Shirjang S, Alizadeh N, Mansoori B, Mahmoodpoor A, Kafil HS, Hojjat-Farsangi M, Yousefi M. Promising immunotherapy: Highlighting cytokine-induced killer cells. J Cell Biochem 2018; 120:8863-8883. [PMID: 30556298 DOI: 10.1002/jcb.28250] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 10/22/2018] [Indexed: 12/20/2022]
Abstract
For many years, cancer therapy has appeared to be a challenging issue for researchers and physicians. By the introduction of novel methods in immunotherapy, the prospect of cancer therapy even more explained than before. Cytokine-induced killer (CIK) cell-based immunotherapy demonstrated to have potentiality in improving clinical outcomes and relieving major side effects of standard treatment options. In addition, given the distinctive features such as high safety, low toxicity effects on healthy cells, numerous clinical trials conducted on CIK cells. Due to the shortcomings that observed in CIK cell immunotherapy alone, arising a tendency to make modifications (combined modality therapy or combination therapy) including the addition of various types of cytokines, genetic engineering, combination with immune checkpoints, and so on. In this review, we have tried to bring forth the latest immunotherapy methods and their overview. We have discussed the combination therapies with CIK cells and the conducted clinical trials. This helps the future studies to use integrated therapies with CIK cells as a promising treatment of many types of cancers.
Collapse
Affiliation(s)
- Solmaz Shirjang
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ata Mahmoodpoor
- Department of Anesthesiology, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammad Hojjat-Farsangi
- Department of Oncology-Pathology, Immune and Gene therapy Lab, Cancer Center Karolinska (CCK), Karolinska University Hospital Solna and Karolinska Institute, Stockholm, Sweden
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
15
|
Vaccine therapy in hematologic malignancies. Blood 2018; 131:2640-2650. [DOI: 10.1182/blood-2017-11-785873] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 02/04/2018] [Indexed: 02/06/2023] Open
Abstract
Abstract
Immune-based therapy has emerged as a paradigm shift in cancer therapy with dramatic responses observed in previously incurable disease. Cancer vaccines are being developed to disrupt tumor-associated tolerance and activate and selectively expand tumor-specific lymphocytes within the native effector cell repertoire while maintaining immune-regulatory protection against autoimmunity. Although individual antigen approaches result in immune response with a suggestion of clinical effect in some settings, broader efficacy may be dependent on presentation of multiple antigens that capture clonal diversity presented in the context of functionally potent antigen-presenting cells. The use of whole cell–based strategies such as dendritic cell/tumor fusions have yielded provocative results in single-arm studies and are currently being explored in multicenter randomized trials. The posttransplant setting is a potentially promising platform for vaccination due to cytoreduction and relative depletion of inhibitory accessory cells fostering greater immune responsiveness. Integration of these efforts with other immunotherapeutic strategies and agents that target the tumor microenvironment is being studied in an effort to generate durable immunologic responses with clinically meaningful impact on disease.
Collapse
|
16
|
Lacher MD, Bauer G, Fury B, Graeve S, Fledderman EL, Petrie TD, Coleal-Bergum DP, Hackett T, Perotti NH, Kong YY, Kwok WW, Wagner JP, Wiseman CL, Williams WV. SV-BR-1-GM, a Clinically Effective GM-CSF-Secreting Breast Cancer Cell Line, Expresses an Immune Signature and Directly Activates CD4 + T Lymphocytes. Front Immunol 2018; 9:776. [PMID: 29867922 PMCID: PMC5962696 DOI: 10.3389/fimmu.2018.00776] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 03/28/2018] [Indexed: 12/18/2022] Open
Abstract
Targeted cancer immunotherapy with irradiated, granulocyte–macrophage colony-stimulating factor (GM-CSF)-secreting, allogeneic cancer cell lines has been an effective approach to reduce tumor burden in several patients. It is generally assumed that to be effective, these cell lines need to express immunogenic antigens coexpressed in patient tumor cells, and antigen-presenting cells need to take up such antigens then present them to patient T cells. We have previously reported that, in a phase I pilot study (ClinicalTrials.gov NCT00095862), a subject with stage IV breast cancer experienced substantial regression of breast, lung, and brain lesions following inoculation with clinical formulations of SV-BR-1-GM, a GM-CSF-secreting breast tumor cell line. To identify diagnostic features permitting the prospective identification of patients likely to benefit from SV-BR-1-GM, we conducted a molecular analysis of the SV-BR-1-GM cell line and of patient-derived blood, as well as a tumor specimen. Compared to normal human breast cells, SV-BR-1-GM cells overexpress genes encoding tumor-associated antigens (TAAs) such as PRAME, a cancer/testis antigen. Curiously, despite its presumptive breast epithelial origin, the cell line expresses major histocompatibility complex (MHC) class II genes (HLA-DRA, HLA-DRB3, HLA-DMA, HLA-DMB), in addition to several other factors known to play immunostimulatory roles. These factors include MHC class I components (B2M, HLA-A, HLA-B), ADA (encoding adenosine deaminase), ADGRE5 (CD97), CD58 (LFA3), CD74 (encoding invariant chain and CLIP), CD83, CXCL8 (IL8), CXCL16, HLA-F, IL6, IL18, and KITLG. Moreover, both SV-BR-1-GM cells and the responding study subject carried an HLA-DRB3*02:02 allele, raising the question of whether SV-BR-1-GM cells can directly present endogenous antigens to T cells, thereby inducing a tumor-directed immune response. In support of this, SV-BR-1-GM cells (which also carry the HLA-DRB3*01:01 allele) treated with yellow fever virus (YFV) envelope (Env) 43–59 peptides reactivated YFV-DRB3*01:01-specific CD4+ T cells. Thus, the partial HLA allele match between SV-BR-1-GM and the clinical responder might have enabled patient T lymphocytes to directly recognize SV-BR-1-GM TAAs as presented on SV-BR-1-GM MHCs. Taken together, our findings are consistent with a potentially unique mechanism of action by which SV-BR-1-GM cells can act as APCs for previously primed CD4+ T cells.
Collapse
Affiliation(s)
| | - Gerhard Bauer
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Brian Fury
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Sanne Graeve
- BriaCell Therapeutics Corp., Berkeley, CA, United States
| | - Emily L Fledderman
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Tye D Petrie
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Dane P Coleal-Bergum
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Tia Hackett
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Nicholas H Perotti
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Ying Y Kong
- Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | - William W Kwok
- Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | | | | | | |
Collapse
|
17
|
Nahas MR, Rosenblatt J, Lazarus HM, Avigan D. Anti-cancer vaccine therapy for hematologic malignancies: An evolving era. Blood Rev 2018; 32:312-325. [PMID: 29475779 DOI: 10.1016/j.blre.2018.02.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 02/08/2018] [Accepted: 02/13/2018] [Indexed: 12/19/2022]
Abstract
The potential promise of therapeutic vaccination as effective therapy for hematologic malignancies is supported by the observation that allogeneic hematopoietic cell transplantation is curative for a subset of patients due to the graft-versus-tumor effect mediated by alloreactive lymphocytes. Tumor vaccines are being explored as a therapeutic strategy to re-educate host immunity to recognize and target malignant cells through the activation and expansion of effector cell populations. Via several mechanisms, tumor cells induce T cell dysfunction and senescence, amplifying and maintaining tumor cell immunosuppressive effects, resulting in failure of clinical trials of tumor vaccines and adoptive T cell therapies. The fundamental premise of successful vaccine design involves the introduction of tumor-associated antigens in the context of effective antigen presentation so that tolerance can be reversed and a productive response can be generated. With the increasing understanding of the role of both the tumor and tumor microenvironment in fostering immune tolerance, vaccine therapy is being explored in the context of immunomodulatory therapies. The most effective strategy may be to use combination therapies such as anti-cancer vaccines with checkpoint blockade to target critical aspects of this environment in an effort to prevent the re-establishment of tumor tolerance while limiting toxicity associated with autoimmunity.
Collapse
Affiliation(s)
- Myrna R Nahas
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| | - Jacalyn Rosenblatt
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Hillard M Lazarus
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - David Avigan
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
18
|
Vaccination with autologous myeloblasts admixed with GM-K562 cells in patients with advanced MDS or AML after allogeneic HSCT. Blood Adv 2017; 1:2269-2279. [PMID: 29296875 DOI: 10.1182/bloodadvances.2017009084] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 10/08/2017] [Indexed: 01/05/2023] Open
Abstract
We report a clinical trial testing vaccination of autologous myeloblasts admixed with granulocyte-macrophage colony-stimulating factor secreting K562 cells after allogeneic hematopoietic stem cell transplantation (HSCT). Patients with myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML) with ≥5% marrow blasts underwent myeloblast collection before HSCT. At approximately day +30, 6 vaccines composed of irradiated autologous myeloblasts mixed with GM-K562 were administered. Tacrolimus-based graft-versus-host disease (GVHD) prophylaxis was not tapered until vaccine completion (∼day 100). Thirty-three patients with AML (25) and MDS (8) enrolled, 16 (48%) had ≥5% marrow blasts at transplantation. The most common vaccine toxicity was injection site reactions. One patient developed severe eosinophilia and died of eosinophilic myocarditis. With a median follow-up of 67 months, cumulative incidence of grade 2-4 acute and chronic GVHD were 24% and 33%, respectively. Relapse and nonrelapse mortality were 48% and 9%, respectively. Progression-free survival (PFS) and overall survival (OS) at 5 years were 39% and 39%. Vaccinated patients who were transplanted with active disease (≥5% marrow blasts) had similar OS and PFS at 5 years compared with vaccinated patients transplanted with <5% marrow blasts (OS, 44% vs 35%, respectively, P = .81; PFS, 44% vs 35%, respectively, P = .34). Postvaccination antibody responses to angiopoietin-2 was associated with superior OS (hazard ratio [HR], 0.43; P = .031) and PFS (HR, 0.5; P = .036). Patients transplanted with active disease had more frequent angiopoeitin-2 antibody responses (62.5% vs 20%, P = .029) than those transplanted in remission. GM-K562/leukemia cell vaccination induces biologic activity, even in patients transplanted with active MDS/AML. This study is registered at www.clinicaltrials.gov as #NCT 00809250.
Collapse
|
19
|
Ventola CL. Cancer Immunotherapy, Part 2: Efficacy, Safety, and Other Clinical Considerations. P & T : A PEER-REVIEWED JOURNAL FOR FORMULARY MANAGEMENT 2017; 42:452-463. [PMID: 28674473 PMCID: PMC5481296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
This article, the second in a series of three, provides an overview of the efficacy and safety of cancer immunotherapies ranging from monoclonal antibodies to vaccines, including additional clinical considerations regarding immune checkpoint blockers.
Collapse
|
20
|
Rafelson WM, Reagan JL, Fast LD, Lim SH. Immunotherapy of elderly acute myeloid leukemia: light at the end of a long tunnel? Leuk Lymphoma 2017; 58:2523-2531. [DOI: 10.1080/10428194.2017.1306646] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- William M. Rafelson
- Division of Hematology and Oncology, Rhode Island Hospital/Brown University Warren Alpert Medical School, Providence, RI, USA
| | - John L. Reagan
- Division of Hematology and Oncology, Rhode Island Hospital/Brown University Warren Alpert Medical School, Providence, RI, USA
| | - Loren D. Fast
- Division of Hematology and Oncology, Rhode Island Hospital/Brown University Warren Alpert Medical School, Providence, RI, USA
| | - Seah H. Lim
- Division of Hematology and Oncology, Rhode Island Hospital/Brown University Warren Alpert Medical School, Providence, RI, USA
| |
Collapse
|
21
|
Hosen N, Maeda T, Hashii Y, Tsuboi A, Nishida S, Nakata J, Oji Y, Oka Y, Sugiyama H. Wilms tumor 1 peptide vaccination after hematopoietic stem cell transplant in leukemia patients. Stem Cell Investig 2016; 3:90. [PMID: 28078270 DOI: 10.21037/sci.2016.11.08] [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: 10/19/2016] [Accepted: 11/07/2016] [Indexed: 01/03/2023]
Abstract
Although the prognosis of leukemia patients after allogeneic hematopoietic stem cell transplantation (HSCT) has greatly improved, relapse is still a major cause of death after HSCT. Cancer vaccines may have the potential to enhance the graft-versus-leukemia (GVL) effect. The post-allogeneic HSCT period provides a unique platform for vaccination, because (I) tumor burden is minimal, (II) lymphopenia allows for rapid expansion of cytotoxic T cells (CTLs), (III) donor-derived CTLs are not exhausted, (IV) inflammation is caused by alloreactions, and (V) the abundance of regulatory T cells is low due to their late recovery. Tumor cell lysates, dendritic cells (DCs), and peptides derived from leukemia-associated antigens (LAAs) have been used as vaccines. Clinical trials with several types of vaccines for post-HSCT patients revealed that the vaccination induced an immunological response and might benefit patients with minimal residual disease; however, the efficacy of this approach must be examined in randomized studies. In addition, it is important to consider the combination of cancer vaccine with checkpoint antibodies, recently shown to be useful in treating leukemia relapse after HSCT.
Collapse
Affiliation(s)
- Naoki Hosen
- Department of Cancer stem cell Biology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tetsuo Maeda
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshiko Hashii
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Akihiro Tsuboi
- Department of Cancer Immunotherapy, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Sumiyuki Nishida
- Department of Respiratory Medicine, Allergy and Rheumatic Diseases, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Jun Nakata
- Department of Cancer Immunotherapy, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yusuke Oji
- Department of Cancer stem cell Biology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshihiro Oka
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Haruo Sugiyama
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
22
|
Mulé MP, Mannis GN, Wood BL, Radich JP, Hwang J, Ramos NR, Andreadis C, Damon L, Logan AC, Martin TG, Hourigan CS. Multigene Measurable Residual Disease Assessment Improves Acute Myeloid Leukemia Relapse Risk Stratification in Autologous Hematopoietic Cell Transplantation. Biol Blood Marrow Transplant 2016; 22:1974-1982. [PMID: 27544285 DOI: 10.1016/j.bbmt.2016.08.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 08/11/2016] [Indexed: 11/25/2022]
Abstract
We report here the largest study to date of adult patients with acute myeloid leukemia (AML) tested for measurable residual disease (MRD) at the time of autologous hematopoietic cell transplantation (auto-HCT). Seventy-two adult patients who underwent transplantation between 2004 and 2013 at a single academic medical center (University of California San Francisco) were eligible for this retrospective study based on availability of cryopreserved granulocyte colony-stimulating factor (GCSF)-mobilized autologous peripheral blood progenitor cell (PBPC) leukapheresis specimens ("autografts"). Autograft MRD was assessed by molecular methods (real-time quantitative PCR [RQ-PCR] for Wilms tumor 1 (WT1) alone or a multigene panel) and by multiparameter flow cytometry (MPFC). WT1 RQ-PCR testing of the autograft had low sensitivity for relapse prediction (14%) and a negative predictive value of 51%. MPFC failed to identify MRD in any of 34 autografts tested. Combinations of molecular MRD assays, however, improved prediction of post-auto-HCT relapse. In multivariate analysis of clinical variables, including age, gender, race, cytogenetic risk category, and CD34+ cell dose, only autograft multigene MRD as assessed by RQ-PCR was statistically significantly associated with relapse. One year after transplantation, only 28% patients with detectable autograft MRD were relapse free, compared with 67% in the MRD-negative cohort. Multigene MRD, while an improvement on other methods tested, was however suboptimal for relapse prediction in unselected patients, with specificity of 83% and sensitivity of 46%. In patients with known chromosomal abnormalities or mutations, however, better predictive value was observed with no relapses observed in MRD-negative patients in the first year after auto-HCT compared with 83% incidence of relapse in the MRD-positive patients (hazard ratio, 12.45; P = .0016). In summary, increased personalization of MRD monitoring by use of a multigene panel improved the ability to risk stratify patients for post-auto-HCT relapse. WT1 RQ-PCR and flow cytometric assessment for AML MRD in autograft samples had limited value for predicting relapse after auto-HCT. We demonstrate that cryopreserved autograft material presents unique challenges for AML MRD testing because of the masking effects of previous GCSF exposure on gene expression and flow cytometry signatures. In the absence of information regarding diagnostic characteristics, sources other than GCSF-stimulated PBSC leukapheresis specimens should be considered as alternatives for MRD testing in AML patients undergoing auto-HCT.
Collapse
Affiliation(s)
- Matthew P Mulé
- Myeloid Malignancies Section, Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Gabriel N Mannis
- Department of Medicine, Division of Hematology and Blood and Marrow Transplantation, University of California, San Francisco, California
| | - Brent L Wood
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | - Jimmy Hwang
- Department of Medicine, Division of Hematology and Blood and Marrow Transplantation, University of California, San Francisco, California
| | - Nestor R Ramos
- Myeloid Malignancies Section, Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Charalambos Andreadis
- Department of Medicine, Division of Hematology and Blood and Marrow Transplantation, University of California, San Francisco, California
| | - Lloyd Damon
- Department of Medicine, Division of Hematology and Blood and Marrow Transplantation, University of California, San Francisco, California
| | - Aaron C Logan
- Department of Medicine, Division of Hematology and Blood and Marrow Transplantation, University of California, San Francisco, California
| | - Thomas G Martin
- Department of Medicine, Division of Hematology and Blood and Marrow Transplantation, University of California, San Francisco, California
| | - Christopher S Hourigan
- Myeloid Malignancies Section, Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
23
|
Rashidi A, Walter RB, Tallman MS, Appelbaum FR, DiPersio JF. Maintenance therapy in acute myeloid leukemia: an evidence-based review of randomized trials. Blood 2016; 128:763-73. [PMID: 27354720 PMCID: PMC4982451 DOI: 10.1182/blood-2016-03-674127] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 06/20/2016] [Indexed: 11/20/2022] Open
Affiliation(s)
- Armin Rashidi
- Section of Bone Marrow Transplantation and Leukemia Program, Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Roland B Walter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA; Division of Hematology, Department of Medicine, and Department of Epidemiology, University of Washington, Seattle, WA
| | - Martin S Tallman
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY; and
| | - Frederick R Appelbaum
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA; Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - John F DiPersio
- Section of Bone Marrow Transplantation and Leukemia Program, Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
24
|
Current status of ex vivo gene therapy for hematological disorders: a review of clinical trials in Japan around the world. Int J Hematol 2016; 104:42-72. [PMID: 27289360 DOI: 10.1007/s12185-016-2030-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 05/22/2016] [Accepted: 05/24/2016] [Indexed: 12/20/2022]
Abstract
Gene therapies are classified into two major categories, namely, in vivo and ex vivo. Clinical trials of human gene therapy began with the ex vivo techniques. Based on the initial successes of gene-therapy clinical trials, these approaches have spread worldwide. The number of gene therapy trials approved worldwide increased gradually starting in 1989, reaching 116 protocols per year in 1999, and a total of 2210 protocols had been approved by 2015. Accumulating clinical evidence has demonstrated the safety and benefits of several types of gene therapy, with the exception of serious adverse events in several clinical trials. These painful experiences were translated backward to basic science, resulting in the development of several new technologies that have influenced the recent development of ex vivo gene therapy in this field. To date, six gene therapies have been approved in a limited number of countries worldwide. In Japan, clinical trials of gene therapy have developed under the strong influence of trials in the US and Europe. Since the initial stages, 50 clinical trials have been approved by the Japanese government. In this review, the history and current status of clinical trials of ex vivo gene therapy for hematological disorders are introduced and discussed.
Collapse
|
25
|
Gulbake A, Jain A, Jain A, Jain A, Jain SK. Insight to drug delivery aspects for colorectal cancer. World J Gastroenterol 2016; 22:582-599. [PMID: 26811609 PMCID: PMC4716061 DOI: 10.3748/wjg.v22.i2.582] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 08/29/2015] [Accepted: 12/01/2015] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer diagnosed worldwide in human beings. Surgery, chemotherapy, radiotherapy and targeted therapies are the conventional four approaches which are currently used for the treatment of CRC. The site specific delivery of chemotherapeutics to their site of action would increase effectiveness with reducing side effects. Targeted oral drug delivery systems based on polysaccharides are being investigated to target and deliver chemotherapeutic and chemopreventive agents directly to colon and rectum. Site-specific drug delivery to colon increases its concentration at the target site, and thus requires a lower dose and hence abridged side effects. Some novel therapies are also briefly discussed in article such as receptor (epidermal growth factor receptor, folate receptor, wheat germ agglutinin, VEGF receptor, hyaluronic acid receptor) based targeting therapy; colon targeted proapoptotic anticancer drug delivery system, gene therapy. Even though good treatment options are available for CRC, the ultimate therapeutic approach is to avert the incidence of CRC. It was also found that CRCs could be prevented by diet and nutrition such as calcium, vitamin D, curcumin, quercetin and fish oil supplements. Immunotherapy and vaccination are used nowadays which are showing better results against CRC.
Collapse
|
26
|
Mannis GN, Martin TG, Damon LE, Logan AC, Olin RL, Flanders MD, Ai WZ, Gaensler KML, Kaplan LD, Sayre PH, Smith CC, Wolf JL, Andreadis C. Long-term outcomes of patients with intermediate-risk acute myeloid leukemia treated with autologous hematopoietic cell transplant in first complete remission. Leuk Lymphoma 2016; 57:1560-6. [PMID: 26490487 DOI: 10.3109/10428194.2015.1088646] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
In 2014, autologous hematopoietic cell transplant (autoHCT) was removed from the National Comprehensive Cancer Network guidelines as a recommended treatment for patients with intermediate-risk AML in first complete remission (CR1). We reviewed the outcomes of all patients with intermediate-risk AML treated with autoHCT in CR1 at our institution. Of 334 patients who underwent autoHCT for AML between 1988 and 2013, 133 patients with intermediate-risk AML in CR1 were identified. Cytogenetics were diploid in 97 (73%). With a median follow-up of 4.1 years (range 0.1-17), median overall survival (OS) is 6.7 years; at 5 years post-transplant, 59% of patients remain alive and 43% remain relapse-free. Forty-eight percent of relapsing patients proceeded to salvage alloHCT. Our findings demonstrate that nearly half of patients with intermediate-risk AML in CR1 achieve sustained remissions, and that salvage alloHCT is feasible in those who relapse. AutoHCT therefore remains a reasonable option for intermediate-risk patients with AML in CR1.
Collapse
Affiliation(s)
- Gabriel N Mannis
- a Department of Medicine, Division of Hematology and Blood and Marrow Transplantation , Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco , San Francisco , CA , USA
| | - Thomas G Martin
- a Department of Medicine, Division of Hematology and Blood and Marrow Transplantation , Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco , San Francisco , CA , USA
| | - Lloyd E Damon
- a Department of Medicine, Division of Hematology and Blood and Marrow Transplantation , Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco , San Francisco , CA , USA
| | - Aaron C Logan
- a Department of Medicine, Division of Hematology and Blood and Marrow Transplantation , Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco , San Francisco , CA , USA
| | - Rebecca L Olin
- a Department of Medicine, Division of Hematology and Blood and Marrow Transplantation , Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco , San Francisco , CA , USA
| | - Michael D Flanders
- a Department of Medicine, Division of Hematology and Blood and Marrow Transplantation , Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco , San Francisco , CA , USA
| | - Weiyun Z Ai
- a Department of Medicine, Division of Hematology and Blood and Marrow Transplantation , Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco , San Francisco , CA , USA
| | - Karin M L Gaensler
- a Department of Medicine, Division of Hematology and Blood and Marrow Transplantation , Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco , San Francisco , CA , USA
| | - Lawrence D Kaplan
- a Department of Medicine, Division of Hematology and Blood and Marrow Transplantation , Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco , San Francisco , CA , USA
| | - Peter H Sayre
- a Department of Medicine, Division of Hematology and Blood and Marrow Transplantation , Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco , San Francisco , CA , USA
| | - Catherine C Smith
- a Department of Medicine, Division of Hematology and Blood and Marrow Transplantation , Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco , San Francisco , CA , USA
| | - Jeffrey L Wolf
- a Department of Medicine, Division of Hematology and Blood and Marrow Transplantation , Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco , San Francisco , CA , USA
| | - Charalambos Andreadis
- a Department of Medicine, Division of Hematology and Blood and Marrow Transplantation , Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco , San Francisco , CA , USA
| |
Collapse
|
27
|
Kapadia CH, Perry JL, Tian S, Luft JC, DeSimone JM. Nanoparticulate immunotherapy for cancer. J Control Release 2015; 219:167-180. [DOI: 10.1016/j.jconrel.2015.09.062] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 09/25/2015] [Accepted: 09/28/2015] [Indexed: 12/11/2022]
|
28
|
Rossmann E, Österborg A, Löfvenberg E, Choudhury A, Forssmann U, von Heydebreck A, Schröder A, Mellstedt H. Mucin 1-specific active cancer immunotherapy with tecemotide (L-BLP25) in patients with multiple myeloma: an exploratory study. Hum Vaccin Immunother 2015; 10:3394-408. [PMID: 25483677 DOI: 10.4161/hv.29918] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Patients (n = 34) with previously untreated, slowly progressive asymptomatic stage I/II multiple myeloma or with stage II/III multiple myeloma in stable response/plateau phase following conventional anti-tumor therapy were immunized repeatedly with the antigen-specific cancer immunotherapeutic agent tecemotide (L-BLP25). Additionally, patients were randomly allocated to either single or multiple low doses of cyclophosphamide to inhibit regulatory T cells (Treg). Immunization with tecemotide resulted in the induction/augmentation of a mucin 1-specific immune response in 47% of patients. The immune responses appeared to involve a Th1-like cellular immune response involving CD4 and CD8 T cells. The rate of immune responses was similar with single versus multiple dosing of cyclophosphamide and in patients with vs. without pre-existing mucin 1 immunity. On-treatment reductions in the slope of M-protein concentration over time (but not fulfilling clinical criteria for responses with conventional anti-tumor agents) were observed in 45% of evaluable patients, predominantly in those without versus with pre-existing mucin 1 immunity and in patients with early stage disease. No differences were seen in patients receiving single or multiple cyclophosphamide dosing. Treatment with tecemotide was generally well tolerated. Repeated vs. single dosing of cyclophosphamide had no impact on Treg numbers and was stopped after a case of fatal encephalitis that was assessed as possibly study-related. Tecemotide immunotherapy induces mucin 1-specific cellular immune responses in a substantial proportion of patients, with preliminary evidence of changes in the M-protein concentration time curve in a subset of patients.
Collapse
Key Words
- ASCI, antigen-specific cancer immunotherapy
- AUC, area under the curve
- Cy, cyclophosphamide
- ELISpot, enzyme-linked immunosorbent spot
- GM-CSF, granulocyte-macrophage colony-stimulating factor
- HR, hazard ratio
- IDA, Immunologic Diagnostic Analysis
- IFN-g, interferon-g
- IL-17, interleukin-17
- IQR, interquartile range
- L-BLP25
- MM, multiple myeloma
- MUC1
- MUC1, mucin 1
- NSCLC, non-small cell lung cancer
- PBMC, peripheral blood mononuclear cell
- TNF-α, tumor necrosis factor-α
- Treg, regulatory T cell
- URR, upper reference range
- immunotherapy
- mucin 1
- multiple myeloma
- tecemotide
Collapse
Affiliation(s)
- Eva Rossmann
- a Karolinska Institute and Karolinska University Hospital ; Stockholm , Sweden
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Le Pogam C, Patel S, Gorombei P, Guerenne L, Krief P, Omidvar N, Tekin N, Bernasconi E, Sicre F, Schlageter MH, Chopin M, Noguera ME, West R, Abu A, Mathews V, Pla M, Fenaux P, Chomienne C, Padua RA. DNA-mediated adjuvant immunotherapy extends survival in two different mouse models of myeloid malignancies. Oncotarget 2015; 6:32494-508. [PMID: 26378812 PMCID: PMC4741708 DOI: 10.18632/oncotarget.5572] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 08/14/2015] [Indexed: 02/06/2023] Open
Abstract
We have previously shown that a specific promyelocytic leukemia-retinoic acid receptor alpha (PML-RARA) DNA vaccine combined with all-trans retinoic acid (ATRA) increases the number of long term survivors with enhanced immune responses in a mouse model of acute promyelocytic leukemia (APL). This study reports the efficacy of a non-specific DNA vaccine, pVAX14Flipper (pVAX14), in both APL and high risk myelodysplastic syndrome (HR-MDS) models. PVAX14 is comprised of novel immunogenic DNA sequences inserted into the pVAX1 therapeutic plasmid. APL mice treated with pVAX14 combined with ATRA had increased survival comparable to that obtained with a specific PML-RARA vaccine. Moreover, the survival advantage correlated with decreased PML-RARA transcript levels and increase in anti-RARA antibody production. In HR-MDS mice, pVAX14 significantly improved survival and reduced biomarkers of leukemic transformation such as phosphorylated mitogen-activated protein/extracellular signal-regulated kinase kinase (MEK) 1. In both preclinical models, pVAX14 vaccine significantly increased interferon gamma (IFNγ) production, memory T-cells (memT), reduced the number of colony forming units (CFU) and increased expression of the adapter molecule signalling to NF-κB, MyD88. These results demonstrate the adjuvant properties of pVAX14 providing thus new approaches to improve clinical outcome in two different models of myeloid malignancies, which may have potential for a broader applicability in other cancers.
Collapse
Affiliation(s)
- Carole Le Pogam
- Unité Mixte de la Recherche de Santé (UMR-S), Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) Unité (U), Paris, France
| | - Satyananda Patel
- Unité Mixte de la Recherche de Santé (UMR-S), Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) Unité (U), Paris, France
| | - Petra Gorombei
- Unité Mixte de la Recherche de Santé (UMR-S), Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) Unité (U), Paris, France
| | - Laura Guerenne
- Unité Mixte de la Recherche de Santé (UMR-S), Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) Unité (U), Paris, France
| | - Patricia Krief
- Unité Mixte de la Recherche de Santé (UMR-S), Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) Unité (U), Paris, France
| | - Nader Omidvar
- Haemotology Department, Cardiff University School of Medicine, Cardiff, UK
| | - Nilgun Tekin
- Biotechnology Institute, Ankara University, Ankara, Turkey
| | - Elena Bernasconi
- Unité Mixte de la Recherche de Santé (UMR-S), Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) Unité (U), Paris, France
| | - Flore Sicre
- Unité Mixte de la Recherche de Santé (UMR-S), Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) Unité (U), Paris, France.,Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Marie-Helene Schlageter
- Unité Mixte de la Recherche de Santé (UMR-S), Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) Unité (U), Paris, France.,Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Martine Chopin
- Département d'Expérimentation Animale, Institut Universitaire d'Hématologie, University Paris Diderot, Paris, France
| | - Maria-Elena Noguera
- Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Robert West
- Welsh Heart Research Institute, Cardiff University School of Medicine, Cardiff, UK
| | - Ansu Abu
- Department of Hematology, Christian Medical College and Hospital, Vellore, India
| | - Vikram Mathews
- Department of Hematology, Christian Medical College and Hospital, Vellore, India
| | - Marika Pla
- Unité Mixte de la Recherche de Santé (UMR-S), Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) Unité (U), Paris, France
| | - Pierre Fenaux
- Unité Mixte de la Recherche de Santé (UMR-S), Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) Unité (U), Paris, France.,Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Christine Chomienne
- Unité Mixte de la Recherche de Santé (UMR-S), Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) Unité (U), Paris, France.,Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Rose Ann Padua
- Unité Mixte de la Recherche de Santé (UMR-S), Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) Unité (U), Paris, France.,Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| |
Collapse
|
30
|
Whole Tumor Antigen Vaccines: Where Are We? Vaccines (Basel) 2015; 3:344-72. [PMID: 26343191 PMCID: PMC4494356 DOI: 10.3390/vaccines3020344] [Citation(s) in RCA: 173] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 04/13/2015] [Accepted: 04/16/2015] [Indexed: 12/19/2022] Open
Abstract
With its vast amount of uncharacterized and characterized T cell epitopes available for activating CD4+ T helper and CD8+ cytotoxic lymphocytes simultaneously, whole tumor antigen represents an attractive alternative source of antigens as compared to tumor-derived peptides and full-length recombinant tumor proteins for dendritic cell (DC)-based immunotherapy. Unlike defined tumor-derived peptides and proteins, whole tumor lysate therapy is applicable to all patients regardless of their HLA type. DCs are essentially the master regulators of immune response, and are the most potent antigen-presenting cell population for priming and activating naïve T cells to target tumors. Because of these unique properties, numerous DC-based immunotherapies have been initiated in the clinics. In this review, we describe the different types of whole tumor antigens that we could use to pulse DCs ex vivo and in vivo. We also discuss the different routes of delivering whole tumor antigens to DCs in vivo and activating them with toll-like receptor agonists.
Collapse
|
31
|
Curran E, Corrales L, Kline J. Targeting the innate immune system as immunotherapy for acute myeloid leukemia. Front Oncol 2015; 5:83. [PMID: 25914882 PMCID: PMC4391043 DOI: 10.3389/fonc.2015.00083] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 03/21/2015] [Indexed: 01/05/2023] Open
Abstract
Because of its disseminated nature and lack of tumor-draining lymph nodes, acute myeloid leukemia (AML) likely employs unique immune evasion strategies as compared to solid malignancies. Targeting these unique mechanisms may result in improved immunotherapeutic approaches. Emerging data suggest that a specific dendritic cell (DC) subset, CD8α DCs, may be responsible for mediating tolerance in AML and thus targeting the innate immune system may be of benefit in this disease. Promising immune targets include the toll-like receptors, calreticulin/CD47, the stimulator of interferon genes pathway, and signal transducer and activator of transcription 3 (STAT3). However, it is becoming clear that compensatory mechanisms may limit the efficacy of these agents alone and thus rationale combinations of immunotherapies are warranted. This review discusses the potential immune evasion strategies in AML, as well as discussion of the promising innate immune targets, both alone and in combination, for this disease.
Collapse
Affiliation(s)
- Emily Curran
- Department of Medicine, University of Chicago , Chicago, IL , USA
| | - Leticia Corrales
- Department of Pathology, University of Chicago , Chicago, IL , USA ; Committee on Immunology, University of Chicago , Chicago, IL , USA
| | - Justin Kline
- Department of Medicine, University of Chicago , Chicago, IL , USA ; Committee on Immunology, University of Chicago , Chicago, IL , USA ; University of Chicago Comprehensive Cancer Center , Chicago, IL , USA
| |
Collapse
|
32
|
Levine BL. Performance-enhancing drugs: design and production of redirected chimeric antigen receptor (CAR) T cells. Cancer Gene Ther 2015; 22:79-84. [DOI: 10.1038/cgt.2015.5] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 12/21/2014] [Accepted: 12/23/2014] [Indexed: 02/02/2023]
|
33
|
Drakes ML, Stiff PJ. Harnessing immunosurveillance: current developments and future directions in cancer immunotherapy. Immunotargets Ther 2014; 3:151-65. [PMID: 27471706 PMCID: PMC4918242 DOI: 10.2147/itt.s37790] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Despite improved methods of cancer detection and disease management over the last few decades, cancer remains a major public health problem in many societies. Conventional therapies, such as chemotherapy, radiation, and surgery, are not usually sufficient to prevent disease recurrence. Therefore, efforts have been focused on developing novel therapies to manage metastatic disease and to prolong disease-free and overall survival, by modulating the immune system to alleviate immunosuppression, and to enhance antitumor immunity. This review discusses protumor mechanisms in patients that circumvent host immunosurveillance, and addresses current immunotherapy modalities designed to target these mechanisms. Given the complexity of cancer immunosuppressive mechanisms, we propose that identification of novel disease biomarkers will drive the development of more targeted immunotherapy. Finally, administration of different classes of immunotherapy in combination regimens, will be the ultimate route to impact low survival rates in advanced cancer patients.
Collapse
Affiliation(s)
- Maureen L Drakes
- Department of Medicine, Division of Hematology and Oncology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Patrick J Stiff
- Department of Medicine, Division of Hematology and Oncology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| |
Collapse
|
34
|
Brusic A, Hainz U, Wadleigh M, Neuberg D, Su M, Canning CM, Deangelo DJ, Stone RM, Lee JS, Mulligan RC, Ritz J, Dranoff G, Sasada T, Wu CJ. Detecting T-cell reactivity to whole cell vaccines: Proof of concept analysis of T-cell response to K562 cell antigens in CML patients. Oncoimmunology 2014; 1:1095-1103. [PMID: 23170257 PMCID: PMC3494623 DOI: 10.4161/onci.20954] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BCR-ABL+ K562 cells hold clinical promise as a component of cancer vaccines, either as bystander cells genetically modified to express immunostimulatory molecules, or as a source of leukemia antigens. To develop a method for detecting T-cell reactivity against K562 cell-derived antigens in patients, we exploited the dendritic cell (DC)-mediated cross-presentation of proteins generated from apoptotic cells. We used UVB irradiation to consistently induce apoptosis of K562 cells, which were then fed to autologous DCs. These DCs were used to both stimulate and detect antigen-specific CD8+ T-cell reactivity. As proof-of-concept, we used cross-presented apoptotic influenza matrix protein-expressing K562 cells to elicit reactivity from matrix protein-reactive T cells. Likewise, we used this assay to detect increased anti-CML antigen T-cell reactivity in CML patients that attained long-lasting clinical remissions following immunotherapy (donor lymphocyte infusion), as well as in 2 of 3 CML patients vaccinated with lethally irradiated K562 cells that were modified to secrete high levels of granulocyte macrophage colony-stimulating factor (GM-CSF). This methodology can be readily adapted to examine the effects of other whole tumor cell-based vaccines, a scenario in which the precise tumor antigens that stimulate immune responses are unknown.
Collapse
Affiliation(s)
- Ana Brusic
- Cancer Vaccine Center; Dana-Farber Cancer Institute; Boston, MA USA ; Monash University; Melbourne, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Isidori A, Salvestrini V, Ciciarello M, Loscocco F, Visani G, Parisi S, Lecciso M, Ocadlikova D, Rossi L, Gabucci E, Clissa C, Curti A. The role of the immunosuppressive microenvironment in acute myeloid leukemia development and treatment. Expert Rev Hematol 2014; 7:807-18. [PMID: 25227702 DOI: 10.1586/17474086.2014.958464] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Functional interplay between acute myeloid leukemia (AML) cells and the bone marrow microenvironment is a distinctive characteristic of this hematological cancer. Indeed, a large body of evidence suggests that proliferation, survival and drug resistance of AML are sustained and modulated by the bone marrow immunosuppressive microenvironment, where both innate and adaptive immune responses are profoundly deregulated. Furthermore, the presence of a number of different immunosuppressive mechanisms results in massive immune deregulation, which causes the eventual escape from natural immune control. Modulating the immune system, as documented by 40 years of stem cell transplantation, may improve survival of AML patients, as the immune system is clearly able to recognize and attack leukemic cells. The understanding of the factors responsible for the escape from immune destruction in AML, which becomes more prominent with disease progression, is necessary for the development of innovative immunotherapeutic treatment modalities in AML.
Collapse
Affiliation(s)
- Alessandro Isidori
- Haematology and Haematopoietic Stem Cell Transplant Center, AORMN Hospital, Via Lombroso, 1, 61122, Pesaro, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Hosen N, Maeda T, Hashii Y, Tsuboi A, Nishida S, Nakata J, Nakae Y, Takashima S, Oji Y, Oka Y, Kumanogoh A, Sugiyama H. Vaccination strategies to improve outcome of hematopoietic stem cell transplant in leukemia patients: early evidence and future prospects. Expert Rev Hematol 2014; 7:671-81. [DOI: 10.1586/17474086.2014.953925] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
37
|
Lymphatic endothelial cells support tumor growth in breast cancer. Sci Rep 2014; 4:5853. [PMID: 25068296 PMCID: PMC4929683 DOI: 10.1038/srep05853] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 07/09/2014] [Indexed: 12/20/2022] Open
Abstract
Tumor lymphatic vessels (LV) serve as a conduit of tumor cell dissemination, due to their leaky nature and secretion of tumor-recruiting factors. Though lymphatic endothelial cells (LEC) lining the LV express distinct factors (also called lymphangiocrine factors), these factors and their roles in the tumor microenvironment are not well understood. Here we employ LEC, microvascular endothelial cells (MEC), and human umbilical vein endothelial cells (HUVEC) cultured in triple-negative MDA-MB-231 tumor-conditioned media (TCM) to determine the factors that may be secreted by various EC in the MDA-MB-231 breast tumor. These factors will serve as endothelium derived signaling molecules in the tumor microenvironment. We co-injected these EC with MDA-MB-231 breast cancer cells into animals and showed that LEC support tumor growth, HUVEC have no significant effect on tumor growth, whereas MEC suppress it. Focusing on LEC-mediated tumor growth, we discovered that TCM-treated LEC (‘tumor-educated LEC') secrete high amounts of EGF and PDGF-BB, compared to normal LEC. LEC-secreted EGF promotes tumor cell proliferation. LEC-secreted PDGF-BB induces pericyte infiltration and angiogenesis. These lymphangiocrine factors may support tumor growth in the tumor microenvironment. This study shows that LV serve a novel role in the tumor microenvironment apart from their classical role as conduits of metastasis.
Collapse
|
38
|
Abstract
Cancer immunotherapy is a promising and effective treatment modality for patients with cancers. Cytokine, anticytokine, and antibody therapies appear to be effective in treating various forms of cancer. The human papillomavirus vaccine is protective for cervical cancer, and this discovery has paved the way to the development of cancer vaccines for other forms of virus-associated cancers such as liver cancer and Merkel cell carcinoma. Clinical trials have demonstrated that adoptive cell therapy using tumor-infiltrating lymphocytes can induce tumor regression in approximately 75% of metastatic melanoma patients, suggesting the possibility of using similar technique to effectively treat breast, lung, and renal cancers in the near future. Besides, genetically engineered T cells transduced with genes encoding specific T cell receptors and chimeric antigen receptors have been shown effective in the treatment of cancer patients. These studies suggest that combination therapies are superior choices in cancer immunotherapy for patients.
Collapse
|
39
|
Lucchini G, Bader P. Hematopoietic stem cell transplantation and immunotherapy for pediatric acute myeloid leukemia: an open challenge. Expert Rev Hematol 2014; 7:291-300. [DOI: 10.1586/17474086.2014.878644] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
40
|
Suzuki K, Aida K, Miyakawa R, Narumi K, Udagawa T, Yoshida T, Ohshima Y, Aoki K. Preimmunization of donor lymphocytes enhances antitumor immunity of autologous hematopoietic stem cell transplantation. Cancer Med 2014; 2:636-45. [PMID: 24403229 PMCID: PMC3892795 DOI: 10.1002/cam4.117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 08/07/2013] [Accepted: 08/08/2013] [Indexed: 01/10/2023] Open
Abstract
Lymphopenia-induced homeostatic proliferation (HP) of T cells following autologous hematopoietic stem cell transplantation (HSCT) skews the T-cell repertoire by engaging tumor-associated antigens (TAAs), leading to an induction of antitumor immunity. Here, as the tumor-reactive lymphocytes preferentially proliferate during the condition of HP, we examined whether the priming of a donor lymphocytes to TAAs could enhance HP-induced antitumor immunity in autologous HSCT recipients. First, to examine whether the tumor-bearing condition of donor influences the antitumor effect of HSCT, the lymphocytes isolated from CT26 tumor-bearing mice were infused into lethally irradiated mice. The growth of tumors was substantially suppressed in the mice that received HSCT from a tumor-bearing donor compared with a naïve donor, suggesting that a fraction of donor lymphocytes from tumor-bearing mice are primed in response to TAAs and remain responsive upon transplantation. We previously reported that type I interferon (IFN) maturates the dendritic cells and promotes the priming of T cells. We then investigated whether the further priming of donor cells by IFN-α can strengthen the antitumor effect of HSCT. The intratumoral IFN-α gene transfer significantly increased the number of IFN-γ-positive lymphocytes in response to CT26 cells but not the syngeneic lymphocytes in donor mice. The infusion of primed donor lymphocytes markedly suppressed the tumor growth in recipient mice, and cured 64% of the treated mice. Autologous HSCT with the infusion of primed donor lymphocytes is a promising strategy to induce an effective antitumor immunity for solid cancers.
Collapse
Affiliation(s)
- Koji Suzuki
- Division of Gene and Immune Medicine, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan; Department of Pediatrics, Fukui University School of Medicine, 23-3 Shimoaizuki, Matsuoka, Yoshida-gun, Fukui, 910-1193, Japan
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Pyzer AR, Avigan DE, Rosenblatt J. Clinical trials of dendritic cell-based cancer vaccines in hematologic malignancies. Hum Vaccin Immunother 2014; 10:3125-31. [PMID: 25625926 PMCID: PMC4514037 DOI: 10.4161/21645515.2014.982993] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 09/17/2014] [Accepted: 10/05/2014] [Indexed: 11/19/2022] Open
Abstract
The potential for the immune system to target hematological malignancies is demonstrated in the allogeneic transplant setting, where durable responses can be achieved. However, allogeneic transplantation is associated with significant morbidity and mortality related to graft versus host disease. Cancer immunotherapy has the capacity to direct a specific cytotoxic immune response against cancer cells, particularly residual cancer cells, in order to reduce the likelihood of disease relapse in a more targeted and tolerated manner. Ex vivo dendritic cells can be primed in various ways to present tumor associated antigen to the immune system, in the context of co-stimulatory molecules, eliciting a tumor specific cytotoxic response in patients. Several approaches to prime dendritic cells and overcome the immunosuppressive microenvironment have been evaluated in pre-clinical and early clinical trials with promising results. In this review, we summarize the clinical data evaluating dendritic cell based vaccines for the treatment of hematological malignancies.
Collapse
Key Words
- AML, Acute Myeloid Leukemia
- ASCT, Autologous Stem Cell Transplant
- Apo-DC, Apoptotic body loaded- dendritic cells
- CML, Chronic Myeloid Leukemia
- CR, Complete response
- CTLA-4, Cytotoxic T-Lymphocyte Antigen 4
- DC/AML, Dendritic cell Acute Myeloid Leukemia fusion vaccine
- DC/MM, Dendritic cell Multiple Myeloma fusion vaccine
- DNA Deoxyribonucleic acid
- FLT-ITD, Fms-like Tyrosine Kinase with Internal Tandem Duplication
- GMCSF, Granulocyte macrophage colony-stimulating factor
- GVHD, Graft vs Host Disease
- HLA-A*2402, Human Leukocyte antigen A*2402
- IFN, Interferon
- IFNg, Interferon gamma
- IL, Interleukin
- Id, Idiotype
- KLH, Keyhole limpet hemocyanin
- MDS, Myelodysplastic syndrome
- MHC, Major histocompatibility complex
- OS, Overall Survival
- PD-1, Programmed death 1
- PD-L1, Programmed death-ligand 1
- PR, Partial response
- PRR, Pathogen recognition receptor
- RNA, Ribonucleic acid
- SCT, Stem cell transplant
- TGFB, Transforming growth factor β
- TNFα, Tumor necrosis factor α
- VEGF, Vascular endothelial growth factor
- VGPR, Very good partial response
- WT-1, Wilm's tumor suppressor gene 1
- cancer
- dendritic cell
- immunotherapy
- leukemia
- mRNA, mRNA
- myeloma
- pDCs, Plasmacytoid Dendritic cell
- trial
- vaccine
Collapse
Affiliation(s)
- Athalia R Pyzer
- Beth Israel Deaconess Medical Center; Harvard Medical School; Boston, MA USA
| | - David E Avigan
- Beth Israel Deaconess Medical Center; Harvard Medical School; Boston, MA USA
| | - Jacalyn Rosenblatt
- Beth Israel Deaconess Medical Center; Harvard Medical School; Boston, MA USA
| |
Collapse
|
42
|
Abstract
Treatment of acute myeloid leukemia (AML) with current chemotherapy regimens is still disappointing, with overall survival rates of ≤40% at 5 years. It is now well established that AML cells can evade the immune system through multiple mechanisms, including the expression of the enzyme indoleamine 2,3 dioxygenase. Immunotherapeutic strategies, including both active, such as vaccination with leukemia-associated antigens, and passive, such as adoptive transfer of allogeneic natural killer cells, may overcome leukemia escape and lead to improved cure. Allogeneic hemopoeitic stem cell transplantation, the most effective treatment of AML, is the best known model of immunotherapy. Following transplant, recipient AML cells are eradicated by donor immune cells through the graft-versus-leukemia (GVL) effect. However, GVL is clinically associated with graft-versus-host disease, the major cause of mortality after transplant. GVL is mediated by donor T cells recognizing either leukemia-associated antigens or minor as well as major histocompatibility antigens. Several innovative strategies have been devised to generate leukemia reactive T cells so as to increase GVL responses with no or little graft-versus-host disease.
Collapse
Affiliation(s)
- Mario Arpinati
- Department of Hematology & Oncological Sciences ‘Seragnoli’, University of Bologna, Italy
| | - Antonio Curti
- Department of Hematology & Oncological Sciences ‘Seragnoli’, University of Bologna, Italy
| |
Collapse
|
43
|
Avigan D, Hari P, Battiwalla M, Bishop MR, Giralt SA, Hardy NM, Kröger N, Wayne AS, Hsu KC. Proceedings from the National Cancer Institute's Second International Workshop on the Biology, Prevention, and Treatment of Relapse after Hematopoietic Stem Cell Transplantation: part II. Autologous Transplantation-novel agents and immunomodulatory strategies. Biol Blood Marrow Transplant 2013; 19:1661-9. [PMID: 24018393 PMCID: PMC3914636 DOI: 10.1016/j.bbmt.2013.08.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Accepted: 08/30/2013] [Indexed: 12/11/2022]
Abstract
In the National Cancer Institute's Second International Workshop on the Biology, Prevention, and Treatment of Relapse after Hematopoietic Stem Cell Transplantation, the Scientific/Educational Session on Autologous Transplantation addressed the role of novel agents and immunomodulatory strategies in management of relapse after autologous hematopoietic stem cell transplantation (AHSCT). Concepts were illustrated through in-depth discussion of multiple myeloma, with broader discussion of areas relevant for relapse of other malignancies as well as in the setting of allogeneic transplantation. Dr. Hari provided an overview of the epidemiology of relapse after AHSCT in multiple myeloma, addressing clinical patterns, management implications, and treatment options at relapse, highlighting the implications of novel therapeutic agents in initial, maintenance, and relapse treatment. Dr. Avigan discussed current concepts in tumor vaccine design, including whole cell and antigen-specific strategies, use of an AHSCT platform to reverse tumor-associated immunosuppression and tolerance, and combining vaccines with immunomodulatory agents to promote establishment of durable antitumor immunity. Dr. Hsu reviewed the immunogenetics of natural killer (NK) cells and general NK biology, the clinical importance of autologous NK activity (eg, lymphoma and neuroblastoma), the impact of existing therapies on promotion of NK cell activity (eg, immunomodulatory drugs, monoclonal antibodies), and strategies for enhancing autologous and allogeneic NK cell effects through NK cell gene profiling.
Collapse
Affiliation(s)
- David Avigan
- Division of Hematology Oncology, Hematologic Malignancies/Bone Marrow Transplant Program, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Putnam AL, Safinia N, Medvec A, Laszkowska M, Wray M, Mintz MA, Trotta E, Szot GL, Liu W, Lares A, Lee K, Laing A, Lechler RI, Riley JL, Bluestone JA, Lombardi G, Tang Q. Clinical grade manufacturing of human alloantigen-reactive regulatory T cells for use in transplantation. Am J Transplant 2013; 13:3010-20. [PMID: 24102808 PMCID: PMC4161737 DOI: 10.1111/ajt.12433] [Citation(s) in RCA: 211] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 07/15/2013] [Accepted: 07/15/2013] [Indexed: 01/25/2023]
Abstract
Regulatory T cell (Treg) therapy has the potential to induce transplantation tolerance so that immunosuppression and associated morbidity can be minimized. Alloantigen-reactive Tregs (arTregs) are more effective at preventing graft rejection than polyclonally expanded Tregs (PolyTregs) in murine models. We have developed a manufacturing process to expand human arTregs in short-term cultures using good manufacturing practice-compliant reagents. This process uses CD40L-activated allogeneic B cells to selectively expand arTregs followed by polyclonal restimulation to increase yield. Tregs expanded 100- to 1600-fold were highly alloantigen reactive and expressed the phenotype of stable Tregs. The alloantigen-expanded Tregs had a diverse TCR repertoire. They were more potent than PolyTregs in vitro and more effective at controlling allograft injuries in vivo in a humanized mouse model.
Collapse
Affiliation(s)
- A. L. Putnam
- UCSF Diabetes Center, University of California, San Francisco, San Francisco CA
| | - N. Safinia
- MRC Centre for Transplantation, King’s College London, London, UK
| | - A. Medvec
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA
| | - M. Laszkowska
- Department of Surgery, University of California, San Francisco, San Francisco CA
| | - M. Wray
- Department of Surgery, University of California, San Francisco, San Francisco CA
| | - M. A. Mintz
- Department of Surgery, University of California, San Francisco, San Francisco CA
| | - E. Trotta
- UCSF Diabetes Center, University of California, San Francisco, San Francisco CA,Department of Surgery, University of California, San Francisco, San Francisco CA
| | - G. L. Szot
- UCSF Diabetes Center, University of California, San Francisco, San Francisco CA,Department of Surgery, University of California, San Francisco, San Francisco CA
| | - W. Liu
- UCSF Diabetes Center, University of California, San Francisco, San Francisco CA
| | - A. Lares
- UCSF Diabetes Center, University of California, San Francisco, San Francisco CA
| | - K. Lee
- Department of Surgery, University of California, San Francisco, San Francisco CA
| | - A. Laing
- MRC Centre for Transplantation, King’s College London, London, UK
| | - R. I. Lechler
- MRC Centre for Transplantation, King’s College London, London, UK
| | - J. L. Riley
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA
| | - J. A. Bluestone
- UCSF Diabetes Center, University of California, San Francisco, San Francisco CA
| | - G. Lombardi
- MRC Centre for Transplantation, King’s College London, London, UK
| | - Q. Tang
- Department of Surgery, University of California, San Francisco, San Francisco CA,Corresponding author: Qizhi Tang,
| |
Collapse
|
45
|
Bot A, Marincola F, Smith KA. Repositioning therapeutic cancer vaccines in the dawning era of potent immune interventions. Expert Rev Vaccines 2013; 12:1219-34. [PMID: 24099049 DOI: 10.1586/14760584.2013.836908] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Based on lessons learned with various immune interventions, this review aims to provide a constructive framework for repositioning therapeutic cancer vaccination. Intensive research throughout the past decade has identified key hurdles interfering with the efficacy of cancer vaccines. The vaccination concept still holds promise if positioned appropriately in minimal residual disease and select early disease stage cancer indications. However, in advanced cancer, it must be integrated with complementary immune interventions to ensure reconstruction of a functional immune repertoire and simultaneous blockade of immune inhibiting mechanisms. Vaccination could render complex and integrative immune interventions simpler, safer and more effective. The near future will witness an explosion of activities in the cancer immunotherapy arena, witnessing a rational repositioning of vaccines rather than their extinction.
Collapse
|
46
|
Induction of high-titer IgG antibodies against multiple leukemia-associated antigens in CML patients with clinical responses to K562/GVAX immunotherapy. Blood Cancer J 2013; 3:e145. [PMID: 24013666 PMCID: PMC3789208 DOI: 10.1038/bcj.2013.44] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 08/07/2013] [Indexed: 01/23/2023] Open
Abstract
The ability to target myeloid leukemia with immunotherapy would represent a significant therapeutic advance. We report here immunological analysis of clinical trials of primary and secondary vaccination with K562/GM-CSF immunotherapy in adult chronic phase chronic myeloid leukemia patients (CML-CP) with suboptimal responses to imatinib mesylate. Using serological analysis of recombinant cDNA expression libraries of K562 with autologous vaccinated patient serum, we have identified 12 novel chronic myeloid leukemia-associated antigens (LAAs). We show that clinical responses following K562/GM-CSF vaccination are associated with induction of high-titer antibody responses to multiple LAAs. We observe markedly discordant patterns of baseline and induced antibody responses in these identically vaccinated patients. No single antigen was recognized in all responses to vaccination. We demonstrate that an additional 'booster' vaccination series can be given safely to those with inadequate responses to initial vaccination, and is associated with more frequent induction of IgG responses to antigens overexpressed in K562 vaccine compared with primary CML-CP. Finally, those with induced immune responses to the same LAAs often shared HLA subtypes and patients with clinical responses following vaccination recognized a partially shared but non-identical spectrum of antigens; both findings have potentially significant implications for cancer vaccine immunotherapy.
Collapse
|
47
|
Udagawa T, Narumi K, Suzuki K, Aida K, Miyakawa R, Ikarashi Y, Makimoto A, Chikaraishi T, Yoshida T, Aoki K. Vascular endothelial growth factor-D-mediated blockade of regulatory T cells within tumors is induced by hematopoietic stem cell transplantation. THE JOURNAL OF IMMUNOLOGY 2013; 191:3440-52. [PMID: 23966628 DOI: 10.4049/jimmunol.1201454] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Lymphopenia-induced homeostatic proliferation of T cells after autologous hematopoietic stem cell transplantation (HSCT) skews the T cell repertoire by engaging tumor-associated Ags, leading to an induction of antitumor immunity. However, how HSCT alters the immunosuppressive microenvironment in the tumors is unknown. In this study, we first analyzed the kinetics of regulatory T cells (Tregs) in the tumors after syngeneic HSCT. Unexpectedly, the frequency of CD4⁺ cells expressing Foxp3 was increased in the spleens, whereas the frequency was clearly decreased in the tumors after HSCT. The origin of reconstituted CD4⁺ and Foxp3⁺ cells in the tumors was mainly from the expansion of transferred splenic T cells. Then, to examine the mechanism of Treg suppression after HSCT, we isolated CD11c⁺ cells from tumors. A large amount of Treg-inhibitory cytokine IL-6 was secreted from the CD11c⁺ cells in the tumors, but not in the spleens in the recipient mice. Furthermore, to understand what factor affects the activity of CD11c⁺ cells in the tumors after HSCT, we analyzed the expression of various cytokines/chemokines with mouse cytokine Ab arrays, and noticed that VEGF-D concentration was increased in the tumors in the early period after HSCT. The CD11c⁺ cells produced IL-6 in response to VEGF-D stimulation, and an administration of VEGF receptor-3 neutralizing Ab significantly suppressed the production of IL-6 from CD11c⁺ cells accompanied with the increase of Tregs in the tumors of HSCT recipients. Autologous HSCT creates an environment that strongly supports the enhancement of antitumor immunity in reconstituted lymphopenic recipients through the suppression of Tregs.
Collapse
Affiliation(s)
- Takeshi Udagawa
- Division of Gene and Immune Medicine, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Burkhardt UE, Hainz U, Stevenson K, Goldstein NR, Pasek M, Naito M, Wu D, Ho VT, Alonso A, Hammond NN, Wong J, Sievers QL, Brusic A, McDonough SM, Zeng W, Perrin A, Brown JR, Canning CM, Koreth J, Cutler C, Armand P, Neuberg D, Lee JS, Antin JH, Mulligan RC, Sasada T, Ritz J, Soiffer RJ, Dranoff G, Alyea EP, Wu CJ. Autologous CLL cell vaccination early after transplant induces leukemia-specific T cells. J Clin Invest 2013; 123:3756-65. [PMID: 23912587 DOI: 10.1172/jci69098] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 05/31/2013] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Patients with advanced hematologic malignancies remain at risk for relapse following reduced-intensity conditioning (RIC) allogeneic hematopoietic stem cell transplantation (allo-HSCT). We conducted a prospective clinical trial to test whether vaccination with whole leukemia cells early after transplantation facilitates the expansion of leukemia-reactive T cells and thereby enhances antitumor immunity. METHODS We enrolled 22 patients with advanced chronic lymphocytic leukemia (CLL), 18 of whom received up to 6 vaccines initiated between days 30 and 45 after transplantation. Each vaccine consisted of irradiated autologous tumor cells admixed with GM-CSF-secreting bystander cells. Serial patient PBMC samples following transplantation were collected, and the impact of vaccination on T cell activity was evaluated. RESULTS At a median follow-up of 2.9 (range, 1-4) years, the estimated 2-year progression-free and overall survival rates of vaccinated subjects were 82% (95% CI, 54%-94%) and 88% (95% CI, 59%-97%), respectively. Although vaccination only had a modest impact on recovering T cell numbers, CD8+ T cells from vaccinated patients consistently reacted against autologous tumor, but not alloantigen-bearing recipient cells with increased secretion of the effector cytokine IFN-γ, unlike T cells from nonvaccinated CLL patients undergoing allo-HSCT. Further analysis confirmed that 17% (range, 13%-33%) of CD8+ T cell clones isolated from 4 vaccinated patients by limiting dilution of bulk tumor-reactive T cells solely reacted against CLL-associated antigens. CONCLUSION Our studies suggest that autologous tumor cell vaccination is an effective strategy to advance long-term leukemia control following allo-HSCT. TRIAL REGISTRATION Clinicaltrials.gov NCT00442130. FUNDING NCI (5R21CA115043-2), NHLBI (5R01HL103532-03), and Leukemia and Lymphoma Society Translational Research Program.
Collapse
Affiliation(s)
- Ute E Burkhardt
- Cancer Vaccine Center, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Ruben JM, Visser LL, Bontkes HJ, Westers TM, Ossenkoppele GJ, de Gruijl TD, van de Loosdrecht AA. Targeting the acute myeloid leukemic stem cell compartment by enhancing tumor cell-based vaccines. Immunotherapy 2013; 5:859-68. [DOI: 10.2217/imt.13.76] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Harvesting the potential of the immune system in order to eradicate (residual) acute myeloid leukemia (AML) cells is the long pursued goal of immunotherapy in AML. Strategies using apoptotic tumor cell vaccines have been explored for many years, without significant clinical improvements. In recent years insight has been gained into the mechanisms activating and interfering with tumor-directed immunity. With the arrival of novel immune-modulating agents allowing for the interference with regulatory molecules and interaction with immune-propelling mechanisms, new doors are opening for increasing vaccination efficacy. Combined with advances in the design of apoptotic tumor-based vaccines, we are on the verge of creating an effective AML vaccine strategy, offering a much needed novel therapeutic option for this devastating disease.
Collapse
Affiliation(s)
- Jurjen M Ruben
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| | - Lindy L Visser
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| | - Hetty J Bontkes
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| | - Theresia M Westers
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| | - Gert J Ossenkoppele
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| | - Arjan A van de Loosdrecht
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands.
| |
Collapse
|
50
|
Ogasawara M. [Leukemia: recent progress in diagnosis and treatment. Topics: IV. Recent topics; 2. Recent advances in cell and immunogene therapy against leukemia]. NIHON NAIKA GAKKAI ZASSHI. THE JOURNAL OF THE JAPANESE SOCIETY OF INTERNAL MEDICINE 2013; 102:1751-8. [PMID: 23947238 DOI: 10.2169/naika.102.1751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|