1
|
Lentiviral Vectors as a Vaccine Platform against Infectious Diseases. Pharmaceutics 2023; 15:pharmaceutics15030846. [PMID: 36986707 PMCID: PMC10053212 DOI: 10.3390/pharmaceutics15030846] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 02/27/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
Lentiviral vectors are among the most effective viral vectors for vaccination. In clear contrast to the reference adenoviral vectors, lentiviral vectors have a high potential for transducing dendritic cells in vivo. Within these cells, which are the most efficient at activating naive T cells, lentiviral vectors induce endogenous expression of transgenic antigens that directly access antigen presentation pathways without the need for external antigen capture or cross-presentation. Lentiviral vectors induce strong, robust, and long-lasting humoral, CD8+ T-cell immunity and effective protection against several infectious diseases. There is no pre-existing immunity to lentiviral vectors in the human population and the very low pro-inflammatory properties of these vectors pave the way for their use in mucosal vaccination. In this review, we have mainly summarized the immunological aspects of lentiviral vectors, their recent optimization to induce CD4+ T cells, and our recent data on lentiviral vector-based vaccination in preclinical models, including prophylaxis against flaviviruses, SARS-CoV-2, and Mycobacterium tuberculosis.
Collapse
|
2
|
Dorff TB, Narayan V, Forman SJ, Zang PD, Fraietta JA, June CH, Haas NB, Priceman SJ. Novel Redirected T-Cell Immunotherapies for Advanced Prostate Cancer. Clin Cancer Res 2022; 28:576-584. [PMID: 34675084 PMCID: PMC8866199 DOI: 10.1158/1078-0432.ccr-21-1483] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/21/2021] [Accepted: 09/13/2021] [Indexed: 01/07/2023]
Abstract
Immunotherapy has failed to achieve durable remissions in advanced prostate cancer patients. More potent T-cell-redirecting strategies may be needed to overcome the immunologically exclusive and suppressive tumor microenvironment. Clinical trials are underway, seeking to define the optimal target for T-cell redirection, such as PSMA, PSCA, or STEAP-1, as well as the optimal strategy, with CAR or bispecific antibodies. As results continue to emerge from these trials, understanding differential toxicity and efficacy of these therapies based on their targets and functional modifications will be key to advancing these promising therapies toward clinical practice. This review provides a unique depth and breadth of perspective regarding the diverse immunotherapy strategies currently under clinical investigation for men with advanced prostate cancer.
Collapse
Affiliation(s)
- Tanya B. Dorff
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Vivek Narayan
- Division of Hematology/Medical Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Stephen J. Forman
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Peter D. Zang
- University of Southern California, Los Angeles, California
| | - Joseph A. Fraietta
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Carl H. June
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Naomi B. Haas
- Division of Hematology/Medical Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Saul J. Priceman
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Comprehensive Cancer Center, Duarte, California
| |
Collapse
|
3
|
Abstract
INTRODUCTION Lentiviral vectors have emerged as powerful vectors for vaccination, due to their high efficiency to transduce dendritic cells and to induce long-lasting humoral immunity, CD8+ T cells, and effective protection in numerous preclinical animal models of infection and oncology. AREAS COVERED Here, we reviewed the literature, highlighting the relevance of lentiviral vectors in vaccinology. We recapitulated both their virological and immunological aspects of lentiviral vectors. We compared lentiviral vectors to the gold standard viral vaccine vectors, i.e. adenoviral vectors, and updated the latest results in lentiviral vector-based vaccination in preclinical models. EXPERT OPINION Lentiviral vectors are non-replicative, negligibly inflammatory, and not targets of preexisting immunity in human populations. These are major characteristics to consider in vaccine development. The potential of lentiviral vectors to transduce non-dividing cells, including dendritic cells, is determinant in their strong immunogenicity. Notably, lentiviral vectors can be engineered to target antigen expression to specific host cells. The very weak inflammatory properties of these vectors allow their use in mucosal vaccination, with particular interest in infectious diseases that affect the lungs or brain, including COVID-19. Recent results in various preclinical models have reinforced the interest of these vectors in prophylaxis against infectious diseases and in onco-immunotherapy.
Collapse
Affiliation(s)
- Min-Wen Ku
- Virology Department, Institut Pasteur-TheraVectys Joint Lab, Paris, France
| | - Pierre Charneau
- Virology Department, Institut Pasteur-TheraVectys Joint Lab, Paris, France
| | - Laleh Majlessi
- Virology Department, Institut Pasteur-TheraVectys Joint Lab, Paris, France
| |
Collapse
|
4
|
Ghaderinia M, Khayamian MA, Abadijoo H, Shalileh S, Faramarzpour M, Zandi A, Simaee H, Abbasvandi F, Esmailinejad MR, Rafizadeh-Tafti S, Jahangiri M, Kordehlachin Y, Ghaffari H, Ansari E, Dabbagh N, Akbari ME, Hoseinpour P, Abdolahad M. Capture-free deactivation of CTCs in the bloodstream; a metastasis suppression method by electrostatic stimulation of the peripheral blood. Biosens Bioelectron 2021; 183:113194. [PMID: 33813209 DOI: 10.1016/j.bios.2021.113194] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/15/2021] [Accepted: 03/18/2021] [Indexed: 12/24/2022]
Abstract
While limited investigations have been reported on CTC elimination and its profits, recently, some new works were reported on detection followed by the destruction of CTCs. Limitations and complications of CTC capturing procedures have highly reduced the chance of selective destruction of CTCs in the bloodstream in the therapeutic guidelines of the patients. Here, we selectively deactivated the invasive function of CTCs during their circulation in the bloodstream by exposing the whole blood to pure positive electrostatic charge stimulation (PPECS). Our treatment suppressed pulmonary metastasis and extended the survival of the mice had been intravenously injected by electrostatically deactivated 4T1 breast cancer CTCs. Moreover, the number of cancerous lung nodules was drastically reduced in the mice injected by treated CTCs in comparison with the non-treated cohort. Evaluating the side effect of the PPECS on the blood components revealed no major effect on the functional properties of the white blood cells, and just a negligible fraction (∼10%) was damaged during this process. This approach does not need any capturing or targeting of CTCs from the blood as it is focused on perturbing the electrical function of negatively-charged tumor cells after being exposed to positive electrostatic charges. Taken together, continuous in-vivo deactivation of CTCs by PPECS with no requirement to complicated capturing protocols may improve the survival of cancer patients.
Collapse
Affiliation(s)
- Mohammadreza Ghaderinia
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran, P.O. Box 14395/515
| | - Mohammad Ali Khayamian
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran, P.O. Box 14395/515
| | - Hamed Abadijoo
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran, P.O. Box 14395/515
| | - Shahriar Shalileh
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran, P.O. Box 14395/515
| | - Mahsa Faramarzpour
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran, P.O. Box 14395/515
| | - Ashkan Zandi
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran, P.O. Box 14395/515
| | - Hossein Simaee
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran, P.O. Box 14395/515; Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, P.O. BOX 15179/64311, Tehran, Iran
| | - Fereshteh Abbasvandi
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran, P.O. Box 14395/515; ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, P.O. BOX 15179/64311, Tehran, Iran
| | - Mohammad Reza Esmailinejad
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran, P.O. Box 14155/6453
| | - Saeed Rafizadeh-Tafti
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran, P.O. Box 14395/515
| | - Mojtaba Jahangiri
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran, P.O. Box 14395/515
| | - Yasin Kordehlachin
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran, P.O. Box 14395/515
| | - Hadi Ghaffari
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran, P.O. Box 14395/515
| | - Ehsan Ansari
- Nano Electronic Center of Excellence, Thin Film and Nano Electronics Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran, P.O. Box 14395/515
| | - Najmeh Dabbagh
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, P.O. BOX 15179/64311, Tehran, Iran
| | - Mohammad Esmaeil Akbari
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, P.O. BOX 15179/64311, Tehran, Iran
| | | | - Mohammad Abdolahad
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran, P.O. Box 14395/515; Cancer Institute, Imam-Khomeini Hospital, Tehran University of Medical Sciences, P.O. BOX 13145-158, Tehran, Iran; UT&TUMS Cancer Electrotechnique Research Center, YAS Hospital, P.O. Box 1598718311, Tehran, Iran.
| |
Collapse
|
5
|
Upadhyay G. Emerging Role of Lymphocyte Antigen-6 Family of Genes in Cancer and Immune Cells. Front Immunol 2019; 10:819. [PMID: 31068932 PMCID: PMC6491625 DOI: 10.3389/fimmu.2019.00819] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 03/27/2019] [Indexed: 12/14/2022] Open
Abstract
Stem Cell Antigen-1 (Sca-1/Ly6A) was the first identified member of the Lymphocyte antigen-6 (Ly6) gene family. Sca-1 serves as a marker of cancer stem cells and tissue resident stem cells in mice. The Sca-1 gene is located on mouse chromosome 15. While a direct homolog of Sca-1 in humans is missing, human chromosome 8—the syntenic region to mouse chromosome 15—harbors several genes containing the characteristic domain known as LU domain. The function of the LU domain in human LY6 gene family is not yet defined. The LY6 gene family proteins are present on human chromosome 6, 8, 11, and 19. The most interesting of these genes are located on chromosome 8q24.3, a frequently amplified locus in human cancer. Human LY6 genes represent novel biomarkers for poor cancer prognosis and are required for cancer progression in addition to playing an important role in immune escape. Although the mechanism associated with these phenotype is not yet clear, it is timely to review the current literature in order to address the critical need for future advancements in this field. This review will summarize recent findings which describe the role of human LY6 genes—LY6D, LY6E, LY6H, LY6K, PSCA, LYPD2, SLURP1, GML, GPIHBP1, and LYNX1; and their orthologs in mice at chromosome 15.
Collapse
Affiliation(s)
- Geeta Upadhyay
- Department of Pathology, John P. Murtha Cancer Center, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
6
|
Abstract
Prostate cancer is the second-most widespread cancer in men worldwide. Treatment choices are limited to prostatectomy, hormonal therapy, and radiotherapy, which commonly have deleterious side effects and vary in their efficacy, depending on the stage of the disease. Among novel experimental strategies, gene therapy holds great promise for the treatment of prostate cancer. However, its use is currently limited by the lack of delivery systems able to selectively deliver the therapeutic genes to the tumors after intravenous administration without major drawbacks. To remediate this problem, a wide range of nonviral delivery approaches have been developed to specifically deliver DNA-based therapeutic agents to their site of action. This review provides an overview of the various nonviral delivery strategies and gene therapy concepts used to deliver therapeutic DNA to prostate cancer cells, and focuses on recent therapeutic advances made so far.
Collapse
Affiliation(s)
- Najla Altwaijry
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK,
| | - Sukrut Somani
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK,
| | - Christine Dufès
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK,
| |
Collapse
|
7
|
Lundstrom K. New frontiers in oncolytic viruses: optimizing and selecting for virus strains with improved efficacy. Biologics 2018; 12:43-60. [PMID: 29445265 PMCID: PMC5810530 DOI: 10.2147/btt.s140114] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Oncolytic viruses have demonstrated selective replication and killing of tumor cells. Different types of oncolytic viruses – adenoviruses, alphaviruses, herpes simplex viruses, Newcastle disease viruses, rhabdoviruses, Coxsackie viruses, and vaccinia viruses – have been applied as either naturally occurring or engineered vectors. Numerous studies in animal-tumor models have demonstrated substantial tumor regression and prolonged survival rates. Moreover, clinical trials have confirmed good safety profiles and therapeutic efficacy for oncolytic viruses. Most encouragingly, the first cancer gene-therapy drug – Gendicine, based on oncolytic adenovirus type 5 – was approved in China. Likewise, a second-generation oncolytic herpes simplex virus-based drug for the treatment of melanoma has been registered in the US and Europe as talimogene laherparepvec.
Collapse
|
8
|
Chen X, Kunda PE, Lin J, Zhou M, Huang J, Zhang H, Liu T. SYK-targeted dendritic cell-mediated cytotoxic T lymphocytes enhance the effect of immunotherapy on retinoblastoma. J Cancer Res Clin Oncol 2018; 144:675-684. [PMID: 29372378 PMCID: PMC5843685 DOI: 10.1007/s00432-018-2584-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 01/15/2018] [Indexed: 12/25/2022]
Abstract
PURPOSE Retinoblastoma (RB) is the most common primary intraocular tumor in children. Chemotherapy is currently the main method of RB treatment. Unfortunately, RB often becomes chemoresistant and turns lethal. Here, we used in vitro cell immunotherapy to explore whether adoptive immunotherapy could be used as a potential treatment for RB. We focused on spleen tyrosine kinase (SYK), which is significantly upregulated in RB cells and serves as a marker for RB cells. METHODS Using lentiviruses, we genetically modified dendritic cells (DCs) to express and present the SYK peptide antigen to cytotoxic T lymphocytes (CTLs) in vitro. We used SYK-negative cell lines (MDA-MB-231, MCF-10A, and hTERT-RPE1) and SYK-positive cell lines (MCF-7 and RB-Y79) to evaluate the specificity and cytotoxicity of DC presented CTLs using FACS, live-cell imaging, and RNA interference. RESULTS The cytotoxicity of CTLs induced by SYK-overexpressing DCs (SYK-DC-CTLs) was enhanced more than three times in SYK-positive cell lines compared with SYK-negative cell lines. DCs primed with SYK could drive CTL cytotoxicity against SYK-positive cell lines but not against SYK-negative cell lines. Moreover, SYK-silenced RB-Y79 cells successfully evaded the cytotoxic attack from SYK-DC-CTLs. However, SYK-DC-CTLs could target SYK overexpressed hTERT-RPE1 cells, suggesting that SYK is a specific antigen for RB. Furthermore, SYK-DC-CTL exhibited specific cytotoxicity against carboplatin-resistant RB-Y79 cells in vitro. CONCLUSIONS Our data showed that SYK could be a potential immunotherapy target mediated by DCs. We propose SYK as a candidate target for treatment of chemoresistant RB.
Collapse
Affiliation(s)
- Xuemei Chen
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Patricia Elena Kunda
- Centro Investigación Medicina Traslacional "Severo Amuchástegui" (CIMETSA), Instituto Universitario Ciencias Biomédicas Córdoba (IUCBC), Córdoba, Argentina
| | - Jianwei Lin
- Shenzhen Key Laboratory for Anti-Ageing and Regenerative Medicine, Health Science Center, Shenzhen University, 3688 Nanhai Avenue, Shenzhen, 518060, Guangdong, China
| | - Meiling Zhou
- Shenzhen Key Laboratory for Anti-Ageing and Regenerative Medicine, Health Science Center, Shenzhen University, 3688 Nanhai Avenue, Shenzhen, 518060, Guangdong, China
- Department of Biotherapy, Shenzhen Luohu People's Hospital, No. 47 Youyi Road, Shenzhen, 518001, Guangdong, China
| | - Jinghan Huang
- Department of Biotherapy, Shenzhen Luohu People's Hospital, No. 47 Youyi Road, Shenzhen, 518001, Guangdong, China
| | - Huqin Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| | - Tao Liu
- Shenzhen Key Laboratory for Anti-Ageing and Regenerative Medicine, Health Science Center, Shenzhen University, 3688 Nanhai Avenue, Shenzhen, 518060, Guangdong, China.
- Department of Biotherapy, Shenzhen Luohu People's Hospital, No. 47 Youyi Road, Shenzhen, 518001, Guangdong, China.
| |
Collapse
|
9
|
Long G, Zhang G, Zhang F, Li M, Ye D, Yang D, Yang Y. Cotransplantation of Mesenchymal Stem Cells and Immature Dendritic Cells Potentiates the Blood Glucose Control of Islet Allografts. BIOMED RESEARCH INTERNATIONAL 2017; 2017:4107943. [PMID: 29410963 PMCID: PMC5749219 DOI: 10.1155/2017/4107943] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 10/23/2017] [Accepted: 11/21/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Transplantation of islets is a promising alternative to treat type 1 diabetes (T1D), but graft rejection is the major obstacle to its application in clinical practice. We evaluated the effects of mesenchymal stem cells (MSCs) and immature dendritic cells (imDCs) on islet transplantation in diabetic model. METHODS The streptozotocin T1D model was established in BABL/c mice. Rat islets were isolated and identified with dithizone (DTZ) staining. MSCs and imDCs were isolated from bone marrow of syngenic mice. Islets, alone or along with MSCs and/or imDCs, were transplanted to the left kidney capsule of diabetic mice. The blood glucose levels and glycosylated hemoglobin levels after transplantation were monitored. RESULTS Cotransplantation significantly decreased blood glucose and glycosylated hemoglobin levels in the diabetes mice. Transplantation of 200 islets + 2 × 105 MSCs + 2 × 105 imDCs could not only restore normal blood glucose levels, but also significantly prolong graft survival for 12.6 ± 3.48 days. CONCLUSIONS Cotransplantation of allogenic islets with imDCs and/or MSCs can significantly promote graft survival, reverse hyperglycemia, and effectively control the glycosylated hemoglobin levels.
Collapse
Affiliation(s)
- Guanghui Long
- Department of Hepatobiliary Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Guangtao Zhang
- Department of Hepatobiliary Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Fangting Zhang
- Center Laboratory, Peking University Shenzhen Hospital, Shenzhen, China
| | - Minghua Li
- Center Laboratory, Peking University Shenzhen Hospital, Shenzhen, China
| | - Dongshuo Ye
- Shenzhen BioScien Pharmaceuticals Co. LTD, Shenzhen, China
| | - Dengke Yang
- Shenzhen BioScien Pharmaceuticals Co. LTD, Shenzhen, China
| | - Yinke Yang
- Shenzhen BioScien Pharmaceuticals Co. LTD, Shenzhen, China
| |
Collapse
|
10
|
Abstract
Gene therapy based on viral vectors has demonstrated steady progress recently, not only in the area of cancers. A multitude of viral vectors has been engineered for both preventive and therapeutic applications. Two main approaches comprise of viral vector-based delivery of toxic or anticancer genes or immunization with anticancer antigens. Tumor growth inhibition and tumor regression have been observed, providing improved survival rates in animal tumor models. Furthermore, vaccine-based cancer immunotherapy has demonstrated both tumor regression and protection against challenges with lethal doses of tumor cells. Several clinical trials with viral vectors have also been conducted. Additionally, viral vector-based cancer drugs have been approved. This review gives an overview of different viral vector systems and their applications in cancer gene therapy.
Collapse
|
11
|
Abstract
Lentiviral vectors (LVs) developed in the past two decades for research and pre-clinical purposes have entered clinical trials with remarkable safety and efficacy performances. Development and clinical testing of LVs for improvement of human immunity showed major advantages in comparison to other viral vector systems. Robust and persisted transduction efficiency of blood cells with LVs, resulted into a broad range of target cells for immune therapeutic approaches: from hematopoietic stem cells and precursor cells for correction of immune deficiencies, up to effector lymphoid and myeloid cells. T cells engineered for expression of chimeric antigen receptors (CARs) or epitope-specific transgenic T cell receptors (TCRs) are in several cancer immune therapy clinical trials worldwide. Development of engineered dendritic cells is primed for clinical trials for cancer and chronic infections. Technological adaptations for ex vivo cell manipulations are here discussed and presented based on properties and uses of the target cell. For future development of off-shelf immune therapies, direct in vivo administration of lentiviral vectors is warranted and intended. Approaches for lentiviral in vivo targeting to maximize immune therapeutic success are discussed.
Collapse
|
12
|
Antigen-presenting cell-targeted lentiviral vectors do not support the development of productive T-cell effector responses: implications for in vivo targeted vaccine delivery. Gene Ther 2017; 24:370-375. [PMID: 28540936 DOI: 10.1038/gt.2017.30] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 02/24/2017] [Accepted: 04/10/2017] [Indexed: 12/22/2022]
Abstract
Targeting transgene expression specifically to antigen-presenting cells (APCs) has been put forward as a promising strategy to direct the immune system towards immunity. We developed the nanobody-display technology to restrict the tropism of lentiviral vectors (LVs) to APCs. However, we observed that immunization with APC-targeted LVs (DC2.1-LVs) did not evoke strong antigen-specific T-cell immunity when compared to immunization with broad tropism LVs (VSV.G-LVs). In this study, we report that VSV.G-LVs are more immunogenic than DC2.1-LVs because they transduce stromal cells, which has a role in activating antigen-specific T cells. Moreover, VSV.G-LVs trigger a pro-inflammatory innate immune response through transduction of APCs and stromal cells, while DC2.1-LVs trigger a type I interferon response with anti-viral capacity. These findings question the rationale of targeting LVs to APCs and argue for the development of VSV.G-LVs with an improved safety profile.
Collapse
|
13
|
Exploratory investigation of PSCA-protein expression in primary breast cancer patients reveals a link to HER2/neu overexpression. Oncotarget 2017; 8:54592-54603. [PMID: 28903367 PMCID: PMC5589606 DOI: 10.18632/oncotarget.17523] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 03/29/2017] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Prostate stem cell antigen (PSCA) has been suggested as biomarker and therapeutic target for prostate cancer. Recent advances showed that PSCA is up-regulated in other cancer entities, such as bladder or pancreatic cancer. However, the clinical relevance of PSCA-expression in breast cancer patients has not yet been established and is therefore addressed by the current study. METHODS PSCA-protein expression was assessed in 405 breast cancer patients, using immunohistochemistry (PSCA antibody MB1) and tissue microarrays. RESULTS PSCA-expression was detected in 94/405 patients (23%) and correlated with unfavorable histopathological grade (p=0.011) and increased Ki67 proliferation index (p=0.006). We observed a strong positive correlation between PSCA-protein expression and HER2/neu receptor status (p<0.001). PSCA did not provide prognostic information in the analyzed cohort. Interestingly, the distribution of PSCA-expression among triple negative patients was comparable to the total population. CONCLUSION We identified a subgroup of PSCA-positive breast cancer patients, which could be amenable for a PSCA-targeted therapy. Moreover, given that we found a strong positive correlation between PSCA- and HER/neu expression, targeting PSCA may provide an alternative therapeutic option in case of trastuzumab resistance.
Collapse
|
14
|
Markov OV, Mironova NL, Shmendel EV, Maslov MA, Zenkova MA. Systemic delivery of complexes of melanoma RNA with mannosylated liposomes activates highly efficient murine melanoma-specific cytotoxic T cells in vivo. Mol Biol 2017. [DOI: 10.1134/s0026893317010137] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
15
|
Mai TJ, Ma R, Li Z, Bi SC. Construction of a fusion plasmid containing the PSCA gene and cytotoxic T-lymphocyte associated antigen-4 (CTLA-4) and its anti-tumor effect in an animal model of prostate cancer. ACTA ACUST UNITED AC 2016; 49:e5620. [PMID: 27783810 PMCID: PMC5089234 DOI: 10.1590/1414-431x20165620] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 09/19/2016] [Indexed: 11/30/2022]
Abstract
Cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) is a negative regulator of T cell activation, which competes with CD28 for B7.1/B7.2 binding, and which has a greater affinity. Fusion of specific antigens to extracellular domain of CTLA4 represents a promising approach to increase the immunogenicity of DNA vaccines. In this study, we evaluated this interesting approach for CTLA4 enhancement on prostate stem cell antigen (PSCA)-specific immune responses and its anti-tumor effects in a prostate cancer mouse model. Consequently, we constructed a DNA vaccine containing the PSCA and the CTLA-4 gene. Vaccination with the CTLA4-fused DNA not only induced a much higher level of anti-PSCA antibody, but also increased PSCA-specific T cell response in mice. To evaluate the anti-tumor efficacy of the plasmids, murine models with PSCA-expressing tumors were generated. After injection of the tumor-bearing mouse model, the plasmid carrying the CTLA4 and PSCA fusion gene showed stronger inhibition of tumor growth than the plasmid expressing PSCA alone. These observations emphasize the potential of the CTLA4-fused DNA vaccine, which could represent a promising approach for tumor immunotherapy.
Collapse
Affiliation(s)
- T J Mai
- Department of Urology, China Meitan General Hospital, Beijing, China
| | - R Ma
- Department of Urology, China Meitan General Hospital, Beijing, China
| | - Z Li
- Department of Urology, China Meitan General Hospital, Beijing, China
| | - S C Bi
- Department of Urology, China Meitan General Hospital, Beijing, China
| |
Collapse
|
16
|
A novel dendritic cell-targeted lentiviral vector, encoding Ag85A-ESAT6 fusion gene of Mycobacterium tuberculosis, could elicit potent cell-mediated immune responses in mice. Mol Immunol 2016; 75:101-11. [DOI: 10.1016/j.molimm.2016.04.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 04/08/2016] [Accepted: 04/28/2016] [Indexed: 01/13/2023]
|
17
|
Abstract
Basic science advances in cancer immunotherapy have resulted in various treatments that have recently shown success in the clinic. Many of these therapies require the insertion of genes into cells to directly kill them or to redirect the host's cells to induce potent immune responses. Other analogous therapies work by modifying effector cells for improved targeting and enhanced killing of tumor cells. Initial studies done using γ-retroviruses were promising, but safety concerns centered on the potential for insertional mutagenesis have highlighted the desire to develop other options for gene delivery. Lentiviral vectors (LVs) have been identified as potentially more effective and safer alternative delivery vehicles. LVs are now in use in clinical trials for many different types of inherited and acquired disorders, including cancer. This review will discuss current knowledge of LVs and the applications of this viral vector-based delivery vehicle to cancer immunotherapy.
Collapse
Affiliation(s)
- Robyn Aa Oldham
- Department of Medical Biophysics, University of Toronto, 27 King's College Circle, Toronto, ON M5S, Canada
| | | | | |
Collapse
|
18
|
Dai S, Zhuo M, Song L, Chen X, Yu Y, Tang Z, Zang G. Dendritic cell-based vaccination with lentiviral vectors encoding ubiquitinated hepatitis B core antigen enhances hepatitis B virus-specific immune responses in vivo. Acta Biochim Biophys Sin (Shanghai) 2015; 47:870-9. [PMID: 26373843 DOI: 10.1093/abbs/gmv093] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 06/30/2015] [Indexed: 12/27/2022] Open
Abstract
The activity of hepatitis B virus (HBV)-specific cytotoxic T lymphocytes (CTLs) plays a predominant role in the clearance of HBV. Dendritic cells (DCs) are key antigen-presenting cells and play an important role in the initiation of immune responses. We previously verified that lentiviral vector encoding ubiquitinated hepatitis B core antigen (LV-Ub-HBcAg) effectively transduced DCs to induce maturation, and the mature DCs efficiently induced T cell polarization to Th1 and generated HBcAg-specific CTLs ex vivo. In this study, HBV-specific immune responses of LV-Ub-HBcAg in BALB/c mice (H-2Kd) were evaluated. It was shown that direct injection of LV-Ub-HBcAg increased the production of cytokines IL-2 and IFN-γ, elicited strong antibody responses, and remarkably generated a high percentage of IFN-γ+CD8+ T cells with HBV-specific CTL responses in BALB/c mice. In addition, direct injection of LV-Ub-HBcAg induced potent anti-HBV immune responses, similar to those elicited by in vitro-transduced DCs. In conclusion, the DC-based therapeutic vaccine LV-Ub-HBcAg elicited specific antibody immune responses and induced robust specific CTL activity in vivo.
Collapse
Affiliation(s)
- Shenglan Dai
- Department of Infectious Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Meng Zhuo
- Department of Infectious Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Linlin Song
- Department of Infectious Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Xiaohua Chen
- Department of Infectious Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Yongsheng Yu
- Department of Infectious Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Zhenghao Tang
- Department of Infectious Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Guoqing Zang
- Department of Infectious Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| |
Collapse
|
19
|
Pros and Cons of Antigen-Presenting Cell Targeted Tumor Vaccines. J Immunol Res 2015; 2015:785634. [PMID: 26583156 PMCID: PMC4637118 DOI: 10.1155/2015/785634] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 08/26/2015] [Accepted: 09/03/2015] [Indexed: 01/08/2023] Open
Abstract
In therapeutic antitumor vaccination, dendritic cells play the leading role since they decide if, how, when, and where a potent antitumor immune response will take place. Since the disentanglement of the complexity and merit of different antigen-presenting cell subtypes, antitumor immunotherapeutic research started to investigate the potential benefit of targeting these subtypes in situ. This review will discuss which antigen-presenting cell subtypes are at play and how they have been targeted and finally question the true meaning of targeting antitumor-based vaccines.
Collapse
|
20
|
Wei SM, Fei JX, Tao F, Pan HL, Shen Q, Wang L, Wu YJ, Zhou L, Zhu SX, Liao WB, Ji H, Xin ZL. Anti-CD27 Antibody Potentiates Antitumor Effect of Dendritic Cell-Based Vaccine in Prostate Cancer-Bearing Mice. Int Surg 2015; 100:155-63. [PMID: 25594656 PMCID: PMC4301282 DOI: 10.9738/intsurg-d-14-00147.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In the current study, we investigated whether anti-CD27 monoclonal antibody can enhance the antitumor efficacy of a dendritic cell-based vaccine in prostate cancer-bearing mice. The overall therapeutic effect of a dendritic cell-based vaccine for prostate cancer remains moderate. A prostate cancer model was established by subcutaneous injection of RM-1 tumor cells into male C57BL/6 mice on day 0. After 4 days, tumor-bearing mice were treated with RM-1 tumor lysate-pulsed dendritic cells (i.e., dendritic cell-based vaccine), anti-CD27 monoclonal antibody, or a combination of RM-1 tumor lysate-pulsed dendritic cells with anti-CD27 monoclonal antibody. Mice were killed at 21 days after tumor cell implantation. Tumor size was measured for assessment of antitumor effect. Spleens were collected for analysis of antitumor immune responses. The antitumor immune responses were evaluated by measuring the proliferation and activity of T cells, which have the ability to kill tumor cells. The combination therapy with RM-1 tumor lysate-pulsed dendritic cells and anti-CD27 antibody significantly enhanced T-cell proliferation and activity, and significantly reduced tumor growth, compared with monotherapy with RM-1 tumor lysate-pulsed dendritic cells or anti-CD27 antibody. Our results suggest that combined treatment can strengthen antitumor efficacy by improving T-cell proliferation and activity.
Collapse
Affiliation(s)
- Si-Ming Wei
- Department of Surgery, Zhejiang Medical College, Hangzhou City, China
- Department of Surgery, Wenzhou Medical University, Wenzhou City, China
| | - Jin-Xuan Fei
- Department of Clinical Medicine, Zhejiang Medical College, Hangzhou City, China
| | - Feng Tao
- Department of Pharmacology, Zhejiang Medical College, Hangzhou City, China
| | - Hang-Li Pan
- Department of Clinical Medicine, Zhejiang Medical College, Hangzhou City, China
| | - Qing Shen
- Department of Surgery, Zhejiang Medical College, Hangzhou City, China
| | - Li Wang
- Department of Clinical Medicine, Zhejiang Medical College, Hangzhou City, China
| | - Yu-Jia Wu
- Department of Clinical Medicine, Zhejiang Medical College, Hangzhou City, China
| | - Li Zhou
- Department of Clinical Medicine, Zhejiang Medical College, Hangzhou City, China
| | - Sheng-Xin Zhu
- Department of Clinical Medicine, Zhejiang Medical College, Hangzhou City, China
| | - Wei-Bin Liao
- Department of Clinical Medicine, Zhejiang Medical College, Hangzhou City, China
| | - Hua Ji
- Department of Basic Medicine, Zhejiang Medical College, Hangzhou City, China
| | - Zhao-Liang Xin
- Department of Surgery, Zhejiang Medical College, Hangzhou City, China
- Department of Surgery, Wenzhou Medical University, Wenzhou City, China
| |
Collapse
|