1
|
Szebeni J. Expanded Spectrum and Increased Incidence of Adverse Events Linked to COVID-19 Genetic Vaccines: New Concepts on Prophylactic Immuno-Gene Therapy, Iatrogenic Orphan Disease, and Platform-Inherent Challenges. Pharmaceutics 2025; 17:450. [PMID: 40284445 PMCID: PMC12029998 DOI: 10.3390/pharmaceutics17040450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/17/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
The mRNA- and DNA-based "genetic" COVID-19 vaccines can induce a broad range of adverse events (AEs), with statistics showing significant variation depending on the timing and data analysis methods used. Focusing only on lipid nanoparticle-enclosed mRNA (mRNA-LNP) vaccines, this review traces the evolution of statistical conclusions on the prevalence of AEs and incidents associated with these vaccines, from initial underestimation of atypical, severe toxicities to recent claims suggesting the possible contribution of COVID-19 vaccinations to the excess deaths observed in many countries over the past few years. Among hundreds of different AEs listed in Pfizer's pharmacovigilance survey, the present analysis categorizes the main symptoms according to organ systems, with nearly all of them being affected. Using data from the US Vaccine Adverse Event Reporting System and a global vaccination dataset, a comparison of the prevalence and incidence rates of AEs induced by genetic versus flu vaccines revealed an average 26-fold increase in AEs with the use of genetic vaccines. The difference is especially pronounced in the case of severe 'Brighton-listed' AEs, which are also observed in COVID-19 and post-COVID conditions. Among these, the increases in incidence rates relative to flu vaccines, given as x-fold rises, were 1152x, 455x, 226x, 218x, 162x, 152x, and 131x for myocarditis, thrombosis, death, myocardial infarction, tachycardia, dyspnea, and hypertension, respectively. The review delineates the concept that genetic vaccines can be regarded as prophylactic immuno-gene therapies and that the observed chronic disabling AEs might be categorized as iatrogenic orphan diseases. It also examines the unique vaccine characteristics that could be causally related to abnormal immune responses which potentially lead to adverse events and complications. These new insights may contribute to improving the safety of this platform technology and assessing the risk/benefit balance of various products.
Collapse
Affiliation(s)
- Janos Szebeni
- Nanomedicine Research and Education Center, Department of Translational Medicine, Semmelweis University, 1089 Budapest, Hungary;
- SeroScience LCC, 1125 Budapest, Hungary
- Translational Nanobioscience Research Center, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
2
|
Gilmour BC, Corthay A, Øynebråten I. High production of IL-12 by human dendritic cells stimulated with combinations of pattern-recognition receptor agonists. NPJ Vaccines 2024; 9:83. [PMID: 38702320 PMCID: PMC11068792 DOI: 10.1038/s41541-024-00869-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 03/29/2024] [Indexed: 05/06/2024] Open
Abstract
The cytokine IL-12p70 is crucial for T helper 1 (Th1) polarization and the generation of type 1 immunity required to fight cancer and pathogens. Therefore, strategies to optimize the production of IL-12p70 by human dendritic cells (DCs) may significantly improve the efficacy of vaccines and immunotherapies. However, the rules governing the production of IL-12p70 remain obscure. Here, we stimulated pattern recognition receptors (PRRs) representing five families of PRRs, to evaluate their ability to elicit high production of IL-12p70 by monocyte-derived DCs. We used ten well-characterized agonists and stimulated DCs in vitro with either single agonists or 27 different combinations. We found that poly(I:C), which engages the RNA-sensing PRRs TLR3 and MDA5, and LPS which stimulates TLR4, were the only agonists that could elicit notable IL-12p70 production when used as single ligands. We identified six different combinations of PRR agonists, all containing either the TLR3/MDA5 agonist poly(I:C) or the TLR7/8 agonist R848, that could synergize to elicit high production of IL-12p70 by human DCs. Five of the six combinations also triggered high production of the antiviral and antitumor cytokine IFNβ. Overall, the tested PRR ligands could be divided into three groups depending on whether they triggered production of both IL-12p70 and IFNβ, only one of the two, or neither. Thus, combinations of PRR agonists were found to increase the production of IL-12p70 by human DCs in a synergistic manner, and we identified six PRR agonist combinations that may represent strong adjuvant candidates, in particular for therapeutic cancer vaccines.
Collapse
Affiliation(s)
- Brian C Gilmour
- Tumor Immunology Lab, Department of Pathology, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | - Alexandre Corthay
- Tumor Immunology Lab, Department of Pathology, Rikshospitalet, Oslo University Hospital, Oslo, Norway
- Hybrid Technology Hub - Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Inger Øynebråten
- Tumor Immunology Lab, Department of Pathology, Rikshospitalet, Oslo University Hospital, Oslo, Norway.
- Hybrid Technology Hub - Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.
| |
Collapse
|
3
|
Fu C, Zhou N, Zhao Y, Duan J, Xu H, Wang Y. Dendritic cells loaded with CD44 + CT-26 colon cell lysate evoke potent antitumor immune responses. Oncol Lett 2019; 18:5897-5904. [PMID: 31788063 PMCID: PMC6865088 DOI: 10.3892/ol.2019.10952] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 09/11/2019] [Indexed: 02/07/2023] Open
Abstract
Increasing evidence supports the concept that cancer stem cells (CSCs) are responsible for cancer progression and metastasis, therapy resistance and relapse. In addition to conventional therapies for colon cancer, the development of immunotherapies targeting cancer stem cells appears to be a promising strategy to suppress tumor recurrence and metastasis. In the present study, dendritic cells (DCs) were pulsed with whole-tumor cell lysates or total RNA of CD44+ colon cancer stem cells (CCSCs) isolated from mouse colon adenocarcinoma CT-26 cell cultures and investigated for their antitumor immunity against CCSCs in vivo and in vitro. In a model of colon adenocarcinoma using BALB/c mice, a sequential reduction in tumor volume and weight was associated with an extended survival in tumor-bearing mice vaccinated with DCs pulsed with RNA or CCSC lysate. In addition, a lactate dehydrogenase assay indicated that cytotoxic T-cells derived from the treated mice exhibited strong cytotoxic activity. Additionally, an enzyme-linked immunosorbent assay revealed that the cytotoxic T-cells of the treated mice released higher levels of interferon-γ against CCSCs compared with those of the control group. In all experiments, the antitumor efficacy of the lysate-pulsed DC-treated and RNA-pulsed DC-treated groups were significantly higher compared with that of the DC-treated and control groups. The results of the present study indicated the potential use of DCs pulsed with cancer stem cell lysates as a potent therapeutic antigen to target CSCs in colon cancer. Additionally, the results provided a rationale for using lysate-pulsed DCs in vivo to eliminate residual tumor deposits in post-operative patients.
Collapse
Affiliation(s)
- Changhao Fu
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ning Zhou
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yuanyuan Zhao
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jinyue Duan
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Hao Xu
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yi Wang
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
4
|
Hossain MK, Nahar K, Donkor O, Apostolopoulos V. Immune-based therapies for metastatic prostate cancer: an update. Immunotherapy 2019; 10:283-298. [PMID: 29421982 DOI: 10.2217/imt-2017-0123] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Prostate cancer (PC) is a common malignancy among elderly males and is noncurable once it becomes metastatic. In recent years, a number of antigen-delivery systems have emerged as viable and promising immunotherapeutic agents against PC. The approval of sipuleucel-T by the US FDA for the treatment of males with asymptomatic or minimally symptomatic castrate resistant PC was a landmark in cancer immunotherapy, making this the first approved immunotherapeutic. A number of vaccines are under clinical investigation, each having its own set of advantages and disadvantages. Here, we discuss the basic technologies underlying these different delivery modes, we discuss the completed and current human clinical trials, as well as the use of vaccines in combination with immune checkpoint inhibitors.
Collapse
Affiliation(s)
| | - Kamrun Nahar
- Vetafarm Pty Ltd, Wagga Wagga, NSW, 2650, Australia
| | - Osaana Donkor
- Centre for Chronic Disease, College of Health & Biomedicine, Victoria University, Melbourne, Australia
| | - Vasso Apostolopoulos
- Centre for Chronic Disease, College of Health & Biomedicine, Victoria University, Melbourne, Australia
| |
Collapse
|
5
|
Bol KF, Schreibelt G, Rabold K, Wculek SK, Schwarze JK, Dzionek A, Teijeira A, Kandalaft LE, Romero P, Coukos G, Neyns B, Sancho D, Melero I, de Vries IJM. The clinical application of cancer immunotherapy based on naturally circulating dendritic cells. J Immunother Cancer 2019. [PMID: 30999964 DOI: 10.1186/s40425-019-0580-] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
Dendritic cells (DCs) can initiate and direct adaptive immune responses. This ability is exploitable in DC vaccination strategies, in which DCs are educated ex vivo to present tumor antigens and are administered into the patient with the aim to induce a tumor-specific immune response. DC vaccination remains a promising approach with the potential to further improve cancer immunotherapy with little or no evidence of treatment-limiting toxicity. However, evidence for objective clinical antitumor activity of DC vaccination is currently limited, hampering the clinical implementation. One possible explanation for this is that the most commonly used monocyte-derived DCs may not be the best source for DC-based immunotherapy. The novel approach to use naturally circulating DCs may be an attractive alternative. In contrast to monocyte-derived DCs, naturally circulating DCs are relatively scarce but do not require extensive culture periods. Thereby, their functional capabilities are preserved, the reproducibility of clinical applications is increased, and the cells are not dysfunctional before injection. In human blood, at least three DC subsets can be distinguished, plasmacytoid DCs, CD141+ and CD1c+ myeloid/conventional DCs, each with distinct functional characteristics. In completed clinical trials, either CD1c+ myeloid DCs or plasmacytoid DCs were administered and showed encouraging immunological and clinical outcomes. Currently, also the combination of CD1c+ myeloid and plasmacytoid DCs as well as the intratumoral use of CD1c+ myeloid DCs is under investigation in the clinic. Isolation and culture strategies for CD141+ myeloid DCs are being developed. Here, we summarize and discuss recent clinical developments and future prospects of natural DC-based immunotherapy.
Collapse
Affiliation(s)
- Kalijn F Bol
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
- Department of Medical Oncology, Radboud university medical centre, Nijmegen, the Netherlands
| | - Gerty Schreibelt
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
| | - Katrin Rabold
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
- Radiotherapy & OncoImmunology Laboratory, Radboud university medical centre, Nijmegen, the Netherlands
| | - Stefanie K Wculek
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares `Carlos III`, Madrid, Spain
| | | | | | - Alvaro Teijeira
- Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Lana E Kandalaft
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - Pedro Romero
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - George Coukos
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - Bart Neyns
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - David Sancho
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares `Carlos III`, Madrid, Spain
| | - Ignacio Melero
- Center for Applied Medical Research, University of Navarra, Pamplona, Spain
- CIBERONC, Madrid, Spain
| | - I Jolanda M de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands.
- Department of Medical Oncology, Radboud university medical centre, Nijmegen, the Netherlands.
| |
Collapse
|
6
|
Bol KF, Schreibelt G, Rabold K, Wculek SK, Schwarze JK, Dzionek A, Teijeira A, Kandalaft LE, Romero P, Coukos G, Neyns B, Sancho D, Melero I, de Vries IJM. The clinical application of cancer immunotherapy based on naturally circulating dendritic cells. J Immunother Cancer 2019; 7:109. [PMID: 30999964 PMCID: PMC6471787 DOI: 10.1186/s40425-019-0580-6] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 03/26/2019] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DCs) can initiate and direct adaptive immune responses. This ability is exploitable in DC vaccination strategies, in which DCs are educated ex vivo to present tumor antigens and are administered into the patient with the aim to induce a tumor-specific immune response. DC vaccination remains a promising approach with the potential to further improve cancer immunotherapy with little or no evidence of treatment-limiting toxicity. However, evidence for objective clinical antitumor activity of DC vaccination is currently limited, hampering the clinical implementation. One possible explanation for this is that the most commonly used monocyte-derived DCs may not be the best source for DC-based immunotherapy. The novel approach to use naturally circulating DCs may be an attractive alternative. In contrast to monocyte-derived DCs, naturally circulating DCs are relatively scarce but do not require extensive culture periods. Thereby, their functional capabilities are preserved, the reproducibility of clinical applications is increased, and the cells are not dysfunctional before injection. In human blood, at least three DC subsets can be distinguished, plasmacytoid DCs, CD141+ and CD1c+ myeloid/conventional DCs, each with distinct functional characteristics. In completed clinical trials, either CD1c+ myeloid DCs or plasmacytoid DCs were administered and showed encouraging immunological and clinical outcomes. Currently, also the combination of CD1c+ myeloid and plasmacytoid DCs as well as the intratumoral use of CD1c+ myeloid DCs is under investigation in the clinic. Isolation and culture strategies for CD141+ myeloid DCs are being developed. Here, we summarize and discuss recent clinical developments and future prospects of natural DC-based immunotherapy.
Collapse
Affiliation(s)
- Kalijn F. Bol
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
- Department of Medical Oncology, Radboud university medical centre, Nijmegen, the Netherlands
| | - Gerty Schreibelt
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
| | - Katrin Rabold
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
- Radiotherapy & OncoImmunology Laboratory, Radboud university medical centre, Nijmegen, the Netherlands
| | - Stefanie K. Wculek
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares `Carlos III`, Madrid, Spain
| | | | | | - Alvaro Teijeira
- Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Lana E. Kandalaft
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - Pedro Romero
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - George Coukos
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - Bart Neyns
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - David Sancho
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares `Carlos III`, Madrid, Spain
| | - Ignacio Melero
- Center for Applied Medical Research, University of Navarra, Pamplona, Spain
- CIBERONC, Madrid, Spain
| | - I. Jolanda M. de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
- Department of Medical Oncology, Radboud university medical centre, Nijmegen, the Netherlands
| |
Collapse
|
7
|
Kyte JA, Fåne A, Pule M, Gaudernack G. Transient redirection of T cells for adoptive cell therapy with telomerase-specific T helper cell receptors isolated from long term survivors after cancer vaccination. Oncoimmunology 2019; 8:e1565236. [PMID: 30906659 DOI: 10.1080/2162402x.2019.1565236] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 11/26/2018] [Accepted: 12/17/2018] [Indexed: 12/22/2022] Open
Abstract
Adoptive cell therapy (ACT) with retargeted T cells has produced remarkable clinical responses against cancer, but also serious toxicity. Telomerase is overexpressed in most cancers, but also expressed in some normal cells, raising safety concerns. We hypothesize that ACT with T-helper cell receptors may overcome tumour tolerance, mobilize host immune cells and induce epitope spreading, with limited toxicity. From long term survivors after cancer vaccination, we have isolated telomerase-specific T cell receptors (TCRs) from T-helper cells. Herein, we report the development of transient retargeting of T cells with mRNA-based TCRs. This strategy allows for safer clinical testing and meaningful dose escalation. DP4 is the most common HLA molecule. We cloned two telomerase-specific, DP4-restricted TCRs into the mRNA expression vector pCIpA102, together with the sorter/marker/suicide gene RQR8. Donor T cells were electroporated with mRNA encoding TCR_RQR8. The results showed that both TCR_RQR8 constructs were expressed in >90% of T cells. The transfected T cells specifically recognized the relevant peptide, as well as naturally processed epitopes from a 177aa telomerase protein fragment, and remained functional for six days. A polyfunctional and Th1-like cytokine profile was observed. The TCRs were functional in both CD4+and CD8+recipient T cells, even though DP4-restricted. The findings demonstrate that the cloned TCRs confer recipient T cells with the desired telomerase-specificity and functionality. Preclinical experiments may provide limited information on the efficacy and toxicity of T-helper TCRs, as these mobilize the host immune system. We therefore intend to use the mRNA-based TCRs for a first-in-man trial.
Collapse
Affiliation(s)
- Jon Amund Kyte
- Department of Oncology, Oslo University Hospital, Oslo, Norway.,Section for Cancer Immunology, Cancer Research Institute, Oslo University Hospital, Oslo, Norway
| | - Anne Fåne
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Martin Pule
- Department of Haematology, Cancer Institute, University College London, London, UK
| | - Gustav Gaudernack
- Section for Cancer Immunology, Cancer Research Institute, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
8
|
Effect of dendritic cell-based immunotherapy on hepatocellular carcinoma: A systematic review and meta-analysis. Cytotherapy 2018; 20:975-989. [PMID: 30072299 DOI: 10.1016/j.jcyt.2018.06.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/17/2018] [Accepted: 06/07/2018] [Indexed: 01/30/2023]
Abstract
BACKGROUND AIMS Dendritic cell (DC)-based immunotherapy has recently been reported frequently in the treatment of hepatocellular carcinoma (HCC); however, its efficacy remains controversial. In this study, we aimed to evaluate the clinical efficacy of DC-based immunotherapy on HCC by conducting a systematic review and meta-analysis. METHODS PubMed, Cochrane Library, Embase and Web of Science were searched to identify clinical trials on DC-based immunotherapy for HCC published up to January 31, 2018. The articles were selected according to pre-established inclusion criteria and methodologic quality, and publication bias were evaluated. RESULTS A total of 1276 cases from 19 clinical trials were included. Compared with traditional treatment, further DC-based therapy enhanced the CD4+ T/CD8+ T ratio (standardized mean difference: 0.68, 95% confidence interval [CI] 0.46-0.89, P < 0.001); increased the 1-year, 18-month and 5-year progression-free survival (PFS) rate and the 1-year, 18-month and 2-year overall survival (OS) rate (relative risk > 1, P < 0.05), prolonged the median PFS time (median survival ratio [MSR]: 1.98, 95% CI: 1.60-2.46, P < 0.001) and median OS time (MSR: 1.72, 95% CI: 1.51-1.96, P < 0.001). Adverse reactions were mild. CONCLUSIONS DC-based therapy not only enhanced anti-tumor immunity, improved the survival rate and prolonged the survival time of HCC patients, but it was also safe. These findings will provide encouraging information for further development of DC-based immunotherapy as an adjuvant treatment for HCC. However, the results must be interpreted with caution because of the small study numbers, publication bias and the various of study designs, pre-treatment and therapeutic processes of DCs.
Collapse
|
9
|
Machlowska J, Maciejewski R, Sitarz R. The Pattern of Signatures in Gastric Cancer Prognosis. Int J Mol Sci 2018; 19:1658. [PMID: 29867026 PMCID: PMC6032410 DOI: 10.3390/ijms19061658] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 05/26/2018] [Accepted: 05/30/2018] [Indexed: 02/05/2023] Open
Abstract
Gastric cancer is one of the most common malignancies worldwide and it is a fourth leading cause of cancer-related death. Carcinogenesis is a multistage disease process specified by the gradual procurement of mutations and epigenetic alterations in the expression of different genes, which finally lead to the occurrence of a malignancy. These genes have diversified roles regarding cancer development. Intracellular pathways are assigned to the expression of different genes, signal transduction, cell-cycle supervision, genomic stability, DNA repair, and cell-fate destination, like apoptosis, senescence. Extracellular pathways embrace tumour invasion, metastasis, angiogenesis. Altered expression patterns, leading the different clinical responses. This review highlights the list of molecular biomarkers that can be used for prognostic purposes and provide information on the likely outcome of the cancer disease in an untreated individual.
Collapse
Affiliation(s)
- Julita Machlowska
- Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland.
| | - Ryszard Maciejewski
- Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland.
| | - Robert Sitarz
- Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland.
- Department of Surgery, St. John's Cancer Center, 20-090 Lublin, Poland.
| |
Collapse
|
10
|
Galati D, Zanotta S. Hematologic neoplasms: Dendritic cells vaccines in motion. Clin Immunol 2017; 183:181-190. [PMID: 28870867 DOI: 10.1016/j.clim.2017.08.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 07/28/2017] [Accepted: 08/29/2017] [Indexed: 12/17/2022]
Abstract
Dendritic cells (DCs) are bone-marrow-derived immune cells accounted for a key role in cancer vaccination as potent antigen-presenting cells within the immune system. Cancer microenvironment can modulate DCs maturation resulting in their accumulation into functional states associated with a reduced antitumor immune response. In this regard, a successful cancer vaccine needs to mount a potent antitumor immune response able to overcome the immunosuppressive tumor milieu. As a consequence, DCs-based approaches are a safe and promising strategy for improving the therapeutic efficacy in hematological malignancies, particularly in combinations with additional treatments. This review summarizes the most significant evidence about the immunotherapeutic strategies performed to target hematologic neoplasms including the tumoral associated antigens (TAA) pulsed on DCs, whole tumor cell vaccines or leukemia-derived DCs.
Collapse
Affiliation(s)
- Domenico Galati
- Hematology-Oncology and Stem-Cell Transplantation Unit, Department of Hematology, National Cancer Institute, Fondazione 'G. Pascale', IRCCS, Via Mariano Semmola 49, 80131 Naples, Italy.
| | - Serena Zanotta
- Hematology-Oncology and Stem-Cell Transplantation Unit, Department of Hematology, National Cancer Institute, Fondazione 'G. Pascale', IRCCS, Via Mariano Semmola 49, 80131 Naples, Italy
| |
Collapse
|
11
|
ALBERTI C. Prostate cancer immunotherapy, particularly in combination with androgen deprivation or radiation treatment. Customized pharmacogenomic approaches to overcome immunotherapy cancer resistance. G Chir 2017; 37:225-235. [PMID: 28098061 PMCID: PMC5256907 DOI: 10.11138/gchir/2016.37.5.225] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Conventional therapeutic approaches for advanced prostate cancer - such as androgen deprivation, chemotherapy, radiation - come up often against lack of effectiveness because of possible arising of correlative cancer cell resistance and/or inadequate anti-tumor immune conditions. Whence the timeliness of resorting to immune-based treatment strategies including either therapeutic vaccination-based active immunotherapy or anti-tumor monoclonal antibody-mediated passive immunotherapy. Particularly attractive, as for research studies and clinical applications, results to be the cytotoxic T-lymphocyte check point blockade by the use of anti-CTLA-4 and PD-1 monoclonal antibodies, particularly when combined with androgen deprivation therapy or radiation. Unlike afore said immune check point inhibitors, both cell-based (by the use of prostate specific antigen carriers autologous dendritic cells or even whole cancer cells) and recombinant viral vector vaccines are able to induce immune-mediated focused killing of specific antigen-presenting prostate cancer cells. Such vaccines, either used alone or concurrently/sequentially combined with above-mentioned conventional therapies, led to generally reach, in the field of various clinical trials, reasonable results particularly as regards the patient's overall survival. Adoptive trasferred T-cells, as adoptive T-cell passive immunotherapy, and monoclonal antibodies against specific antigen-endowed prostate cancer cells can improve immune micro-environmental conditions. On the basis of a preliminary survey about various immunotherapy strategies, are here also outlined their effects when combined with androgen deprivation therapy or radiation. What's more, as regard the immune-based treatment effectiveness, it has to be pointed out that suitable personalized epigenetic/gene profile-achieved pharmacogenomic approaches to target identified gene aberrations, may lead to overcome - as well as for conventional therapies - possible prostate cancer resistance to immunotherapy.
Collapse
|
12
|
Abraham RS, Mitchell DA. Gene-modified dendritic cell vaccines for cancer. Cytotherapy 2017; 18:1446-1455. [PMID: 27745604 DOI: 10.1016/j.jcyt.2016.09.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 09/16/2016] [Indexed: 12/13/2022]
Abstract
Dendritic cell (DC) vaccines are an immunotherapeutic approach to cancer treatment that use the antigen-presentation machinery of DCs to activate an endogenous anti-tumor response. In this treatment strategy, DCs are cultured ex vivo, exposed to tumor antigens and administered to the patient. The ex vivo culturing provides a unique and powerful opportunity to modify and enhance the DCs. As such, a variety of genetic engineering approaches have been employed to optimize DC vaccines, including the introduction of messenger RNA and small interfering RNA, viral gene transduction, and even fusion with whole tumor cells. In general, these modifications aim to improve targeting, enhance immunogenicity, and reduce susceptibility to the immunosuppressive tumor microenvironment. It has been demonstrated that several of these modifications can be employed in tandem, allowing for fine-tuning and optimization of the DC vaccine across multiple metrics. Thus, the application of genetic engineering techniques to the dendritic cell vaccine platform has the potential to greatly enhance its efficacy in the clinic.
Collapse
Affiliation(s)
- Rebecca S Abraham
- UF Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Department of Neurosurgery, University of Florida, Gainesville, FL 32605
| | - Duane A Mitchell
- UF Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Department of Neurosurgery, University of Florida, Gainesville, FL 32605.
| |
Collapse
|
13
|
Creusot RJ, Battaglia M, Roncarolo MG, Fathman CG. Concise Review: Cell-Based Therapies and Other Non-Traditional Approaches for Type 1 Diabetes. Stem Cells 2016; 34:809-19. [PMID: 26840009 PMCID: PMC5021120 DOI: 10.1002/stem.2290] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 12/07/2015] [Indexed: 01/01/2023]
Abstract
The evolution of Type 1 diabetes (T1D) therapy has been marked by consecutive shifts, from insulin replacement to immunosuppressive drugs and targeted biologics (following the understanding that T1D is an autoimmune disease), and to more disease‐specific or patient‐oriented approaches such as antigen‐specific and cell‐based therapies, with a goal to provide efficacy, safety, and long‐term protection. At the same time, another important paradigm shift from treatment of new onset T1D patients to prevention in high‐risk individuals has taken place, based on the hypothesis that therapeutic approaches deemed sufficiently safe may show better efficacy if applied early enough to maintain endogenous β cell function, a concept supported by many preclinical studies. This new strategy has been made possible by capitalizing on a variety of biomarkers that can more reliably estimate the risk and rate of progression of the disease. More advanced (“omic”‐based) biomarkers that also shed light on the underlying contributors of disease for each individual will be helpful to guide the choice of the most appropriate therapies, or combinations thereof. In this review, we present current efforts to stratify patients according to biomarkers and current alternatives to conventional drug‐based therapies for T1D, with a special emphasis on cell‐based therapies, their status in the clinic and potential for treatment and/or prevention. Stem Cells2016;34:809–819
Collapse
Affiliation(s)
- Remi J Creusot
- Department of Medicine, Columbia Center for Translational Immunology and Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, USA
| | - Manuela Battaglia
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria-Grazia Roncarolo
- Division of Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine Stanford, CA, USA
| | - C Garrison Fathman
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine Stanford, CA, USA
| |
Collapse
|
14
|
Galluzzi L, Senovilla L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial watch: Dendritic cell-based interventions for cancer therapy. Oncoimmunology 2014; 1:1111-1134. [PMID: 23170259 PMCID: PMC3494625 DOI: 10.4161/onci.21494] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Dendritic cells (DCs) occupy a central position in the immune system, orchestrating a wide repertoire of responses that span from the development of self-tolerance to the elicitation of potent cellular and humoral immunity. Accordingly, DCs are involved in the etiology of conditions as diverse as infectious diseases, allergic and autoimmune disorders, graft rejection and cancer. During the last decade, several methods have been developed to load DCs with tumor-associated antigens, ex vivo or in vivo, in the attempt to use them as therapeutic anticancer vaccines that would elicit clinically relevant immune responses. While this has not always been the case, several clinical studies have demonstrated that DC-based anticancer vaccines are capable of activating tumor-specific immune responses that increase overall survival, at least in a subset of patients. In 2010, this branch of clinical research has culminated with the approval by FDA of a DC-based therapeutic vaccine (sipuleucel-T, Provenge®) for use in patients with asymptomatic or minimally symptomatic metastatic hormone-refractory prostate cancer. Intense research efforts are currently dedicated to the identification of the immunological features of patients that best respond to DC-based anticancer vaccines. This knowledge may indeed lead to personalized combination strategies that would extend the benefit of DC-based immunotherapy to a larger patient population. In addition, widespread enthusiasm has been generated by the results of the first clinical trials based on in vivo DC targeting, an approach that holds great promises for the future of DC-based immunotherapy. In this Trial Watch, we will summarize the results of recently completed clinical trials and discuss the progress of ongoing studies that have evaluated/are evaluating DC-based interventions for cancer therapy.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France ; Institut Gustave Roussy; Villejuif, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Westdorp H, Sköld AE, Snijer BA, Franik S, Mulder SF, Major PP, Foley R, Gerritsen WR, de Vries IJM. Immunotherapy for prostate cancer: lessons from responses to tumor-associated antigens. Front Immunol 2014; 5:191. [PMID: 24834066 PMCID: PMC4018526 DOI: 10.3389/fimmu.2014.00191] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 04/17/2014] [Indexed: 12/15/2022] Open
Abstract
Prostate cancer (PCa) is the most common cancer in men and the second most common cause of cancer-related death in men. In recent years, novel therapeutic options for PCa have been developed and studied extensively in clinical trials. Sipuleucel-T is the first cell-based immunotherapeutic vaccine for treatment of cancer. This vaccine consists of autologous mononuclear cells stimulated and loaded with an immunostimulatory fusion protein containing the prostate tumor antigen prostate acid posphatase. The choice of antigen might be key for the efficiency of cell-based immunotherapy. Depending on the treatment strategy, target antigens should be immunogenic, abundantly expressed by tumor cells, and preferably functionally important for the tumor to prevent loss of antigen expression. Autoimmune responses have been reported against several antigens expressed in the prostate, indicating that PCa is a suitable target for immunotherapy. In this review, we will discuss PCa antigens that exhibit immunogenic features and/or have been targeted in immunotherapeutic settings with promising results, and we highlight the hurdles and opportunities for cancer immunotherapy.
Collapse
Affiliation(s)
- Harm Westdorp
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , Netherlands ; Department of Medical Oncology, Radboud University Medical Center , Nijmegen , Netherlands
| | - Annette E Sköld
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , Netherlands
| | - Berit A Snijer
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , Netherlands
| | - Sebastian Franik
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , Netherlands
| | - Sasja F Mulder
- Department of Medical Oncology, Radboud University Medical Center , Nijmegen , Netherlands
| | - Pierre P Major
- Juravinski Hospital and Cancer Centre , Hamilton, ON , Canada
| | - Ronan Foley
- Juravinski Hospital and Cancer Centre , Hamilton, ON , Canada
| | - Winald R Gerritsen
- Department of Medical Oncology, Radboud University Medical Center , Nijmegen , Netherlands
| | - I Jolanda M de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , Netherlands ; Department of Medical Oncology, Radboud University Medical Center , Nijmegen , Netherlands
| |
Collapse
|
16
|
Wimmers F, Schreibelt G, Sköld AE, Figdor CG, De Vries IJM. Paradigm Shift in Dendritic Cell-Based Immunotherapy: From in vitro Generated Monocyte-Derived DCs to Naturally Circulating DC Subsets. Front Immunol 2014; 5:165. [PMID: 24782868 PMCID: PMC3990057 DOI: 10.3389/fimmu.2014.00165] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 03/28/2014] [Indexed: 12/31/2022] Open
Abstract
Dendritic cell (DC)-based immunotherapy employs the patients’ immune system to fight neoplastic lesions spread over the entire body. This makes it an important therapy option for patients suffering from metastatic melanoma, which is often resistant to chemotherapy. However, conventional cellular vaccination approaches, based on monocyte-derived DCs (moDCs), only achieved modest response rates despite continued optimization of various vaccination parameters. In addition, the generation of moDCs requires extensive ex vivo culturing conceivably hampering the immunogenicity of the vaccine. Recent studies, thus, focused on vaccines that make use of primary DCs. Though rare in the blood, these naturally circulating DCs can be readily isolated and activated thereby circumventing lengthy ex vivo culture periods. The first clinical trials not only showed increased survival rates but also the induction of diversified anti-cancer immune responses. Upcoming treatment paradigms aim to include several primary DC subsets in a single vaccine as pre-clinical studies identified synergistic effects between various antigen-presenting cells.
Collapse
Affiliation(s)
- Florian Wimmers
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , Netherlands
| | - Gerty Schreibelt
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , Netherlands
| | - Annette E Sköld
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , Netherlands
| | - I Jolanda M De Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , Netherlands ; Department of Medical Oncology, Radboud University Medical Center , Nijmegen , Netherlands
| |
Collapse
|
17
|
Komine H, Kuhn L, Matsushita N, Mulé JJ, Pilon-Thomas S. Examination of MARCO activity on dendritic cell phenotype and function using a gene knockout mouse. PLoS One 2013; 8:e67795. [PMID: 23840879 PMCID: PMC3698187 DOI: 10.1371/journal.pone.0067795] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 05/17/2013] [Indexed: 01/03/2023] Open
Abstract
We have reported the upregulation of MARCO, a member of the class A scavenger receptor family, on the surface of murine and human dendritic cells (DCs) pulsed with tumor lysates. Exposure of murine tumor lysate-pulsed DCs to an anti-MARCO antibody led to loss of dendritic-like processes and enhanced migratory capacity. In this study, we have further examined the biological and therapeutic implications of MARCO expression by DCs. DCs generated from the bone marrow (bm) of MARCO knockout (MARCO-/-) mice were phenotypically similar to DCs generated from the bm of wild-type mice and produced normal levels of IL-12 and TNF-α when exposed to LPS. MARCO-/- DCs demonstrated enhanced migratory capacity in response to CCL-21 in vitro. After subcutaneous injection into mice, MARCO-/- TP-DCs migrated more efficiently to the draining lymph node leading to enhanced generation of tumor-specific IFN-γ producing T cells and improved tumor regression and survival in B16 melanoma-bearing mice. These results support targeting MARCO on the surface of DCs to improve trafficking and induction of anti-tumor immunity.
Collapse
Affiliation(s)
- Hiroshi Komine
- Immunology Program, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Lisa Kuhn
- Immunology Program, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Norimasa Matsushita
- Immunology Program, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - James J. Mulé
- Immunology Program, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
- Cutaneous Oncology Program, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Shari Pilon-Thomas
- Immunology Program, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
- Cutaneous Oncology Program, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
- * E-mail:
| |
Collapse
|
18
|
Abstract
Ex vivo activated B cells are an alternative source of antigen presenting cells (APC). However, the ability of ex vivo activated B cells to function as potent APCs has been a concern especially when compared to dendritic cells (DC). Herein, we introduce a strategy to modulate antigen presentation and immune stimulation functions of activated B cells by co-transfection with multiple mRNAs encoding costimulatory molecules (OX40L, 4-1BBL, and CD80), cytokines (IL-12p35 and IL-12p40) and antigen. These activated B cells modified to express immune stimulatory molecules can be a potent alternative to DCs in immunotherapy.
Collapse
|
19
|
Jørgensen JAL, Longva AS, Hovig E, Bøe SL. Evaluation of Biodegradable Peptide Carriers for Light-Directed Targeting. Nucleic Acid Ther 2013; 23:131-9. [DOI: 10.1089/nat.2012.0403] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
| | - Ane Sager Longva
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo, Norway
| | - Eivind Hovig
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo, Norway
| | - Sigurd Leinæs Bøe
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo, Norway
| |
Collapse
|
20
|
Yu Z, Qian J, Wu J, Gao J, Zhang M. Allogeneic mRNA-based electrotransfection of autologous dendritic cells and specific antitumor effects against osteosarcoma in rats. Med Oncol 2012; 29:3440-8. [PMID: 22843292 DOI: 10.1007/s12032-012-0312-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 07/16/2012] [Indexed: 10/28/2022]
Abstract
Vaccination with dendritic cells (DCs) transfected with tumor-derived mRNA antigen has emerged as a promising strategy for generating protective immunity in mammals. However, the integration of allogeneic osteosarcoma mRNA and autologous DCs has not been fully examined. This study was designed to investigate the antitumor effects of tumor vaccine produced by autologous DCs transfected of allogeneic osteosarcoma mRNA through electroporation in tumor-bearing rats model. In the present study, extraction of Wistar rat tumor mRNA was performed as a two-step procedure. First, total RNA was extracted by use of Trizol; then, mRNA purification was performed by use of polyT-coated magnetic beads. Then, we transfected the allogeneic-derived tumor mRNA to Sprague-Dawley (SD) rat bone marrow-derived DCs through electroporation. The tumor vaccine was applied to tumor-bearing rats model, and the specific antitumor effects of the tumor vaccine were observed. The immunization using autologous DCs electrotransfected with allogeneic osteosarcoma total RNA induced specific CTL responses, which were statistically significant (P < 0.05), and the cytotoxic activity was confirmed in cold target inhibition assays and using mAbs blocking MHC class I molecules. In in vivo experiments, 70 % of the rats immunized with allogeneic osteosarcoma RNA transfected to DCs were typically able to reject tumor challenge and remained tumor-free. Vaccinated survivors developed long immunological memory and were able to reject a subsequent rechallenge with the same tumor cells but not a syngeneic unrelated tumor line. In the present study, we demonstrated that allogeneic tumor mRNA isolated from rat osteosarcoma cell line could be applied to produce tumor vaccine inducing specific antitumor effects, especially in DC-based immunotherapy strategy. This study also provides the foundations for an effective and broadly applicable treatment to a wide range of cancer indications for which tumor-associated antigens have not been identified.
Collapse
Affiliation(s)
- Zhe Yu
- Center of Orthopedic Surgery, Orthopedics Oncology Institute of Chinese PLA, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi, People's Republic of China.
| | | | | | | | | |
Collapse
|
21
|
Zohra FT, Maitani Y, Akaike T. mRNA delivery through fibronectin associated liposome-apatite particles: a new approach for enhanced mRNA transfection to mammalian cell. Biol Pharm Bull 2012; 35:111-5. [PMID: 22223346 DOI: 10.1248/bpb.35.111] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
It was believed for a long time that mRNA is very unstable, and can not be used for therapeutic purposes. In the last decade, however, many research groups proved its transfection feasibility along with advantages and applications. Our investigation is aimed at establishing a potent and efficient mRNA delivery system. We previously reported that an inorganic-organic hybrid carrier by exploiting the advantages of inorganic nano apatite particles onto organic carrier DOTAP {N-[1-(2,3-dioleoloxy)propyl]-N,N,N-trimethyl ammonium chloride} and showed potential effect of carbonate apatite particles on each of the mRNA delivery steps in dividing and non-dividing cell. Here, we report on the development of a more efficient mRNA carrier by complexing ECM protein, fibronectin with the DOTAP-apatite carrier. The carrier showed enhanced uptake of luciferase mRNA both qualitatively and quantitatively. Accelerated cellular endocytosis rate was evaluated using labeled endosome. Finally expression of lucifearse mRNA was higher for fibronectin complexed carrier in compared to the uncoated one.
Collapse
Affiliation(s)
- Fatema Tuj Zohra
- Department of Biomolecular Engineering, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226–8501, Japan
| | | | | |
Collapse
|
22
|
Etschel JK, Hückelhoven AG, Hofmann C, Zitzelsberger K, Maurer K, Bergmann S, Mueller-Schmucker SM, Wittmann J, Spriewald BM, Dörrie J, Schaft N, Harrer T. HIV-1 mRNA electroporation of PBMC: A simple and efficient method to monitor T-cell responses against autologous HIV-1 in HIV-1-infected patients. J Immunol Methods 2012; 380:40-55. [DOI: 10.1016/j.jim.2012.03.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 03/23/2012] [Accepted: 03/23/2012] [Indexed: 10/28/2022]
|
23
|
Abstract
The recent approval of Sipuleucel-T (Dendreon, Seattle, WA) from the Food and Drug Administration for the treatment of men with asymptomatic or minimally symptomatic castrate-resistant prostate cancer was a landmark in cancer immunotherapy, making this the first cancer "vaccine" approved for use in a treatment setting. This approval has led to renewed interest in cancer vaccines and to the recognition that prostate cancer represents an immunologically sensitive disease. At the current time, several vaccine approaches are under clinical investigation. These include viral vectors, antigen-loaded dendritic cells, and DNA vaccines. Each approach has its own set of advantages and disadvantages. This review will introduce the basic technology underlying these different vaccines and briefly discuss completed and ongoing clinical trials. As a great number of prostate cancer vaccines have been investigated in both preclinical and clinical settings, we will focus primarily on vaccines that are currently in clinical trials, as ascertained by a recent inquiry of the clinical trials database, www.clinicaltrials.gov.
Collapse
|
24
|
Lesterhuis WJ, de Vries IJM, Schreibelt G, Lambeck AJ, Aarntzen EH, Jacobs JF, Scharenborg NM, van de Rakt MW, de Boer AJ, Croockewit S, van Rossum MM, Mus R, Oyen WJ, Boerman OC, Lucas S, Adema GJ, Punt CJ, Figdor CG. Route of Administration Modulates the Induction of Dendritic Cell Vaccine–Induced Antigen-Specific T Cells in Advanced Melanoma Patients. Clin Cancer Res 2011; 17:5725-35. [DOI: 10.1158/1078-0432.ccr-11-1261] [Citation(s) in RCA: 131] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
25
|
Shurin MR, Gregory M, Morris JC, Malyguine AM. Genetically modified dendritic cells in cancer immunotherapy: a better tomorrow? Expert Opin Biol Ther 2011; 10:1539-53. [PMID: 20955111 DOI: 10.1517/14712598.2010.526105] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
IMPORTANCE OF THE FIELD Dendritic cells (DC) are powerful antigen-presenting cells that induce and maintain primary cytotoxic T lymphocyte (CTL) responses directed against tumor antigens. Consequently, there has been much interest in their application as antitumor vaccines. AREAS COVERED IN THIS REVIEW A large number of DC-based vaccine trials targeting a variety of cancers have been conducted; however, the rate of reported clinically significant responses remains low. Modification of DC to express tumor antigens or immunostimulatory molecules through the transfer of genes or mRNA transfection offers a logical alternative with potential advantages over peptide- or protein antigen-loaded DC. In this article, we review the current results and future prospects for genetically modified DC vaccines for the treatment of cancer. WHAT THE READER WILL GAIN Genetically-modified dendritic cell-based vaccines represent a powerful tool for cancer therapy. Numerous preclinical and clinical studies have demonstrated the potential of dendritic cell vaccines alone or in combination with other therapeutic modalities. TAKE HOME MESSAGE Genetically modified DC-based anti-cancer vaccination holds promise, perhaps being best employed in the adjuvant setting with minimal residual disease after primary therapy, or in combination with other antitumor or immune-enhancing therapies.
Collapse
Affiliation(s)
- Michael R Shurin
- Department of Pathology and Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | | | | |
Collapse
|
26
|
Tavernier G, Andries O, Demeester J, Sanders NN, De Smedt SC, Rejman J. mRNA as gene therapeutic: How to control protein expression. J Control Release 2011; 150:238-47. [DOI: 10.1016/j.jconrel.2010.10.020] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Accepted: 10/13/2010] [Indexed: 10/18/2022]
|
27
|
Zhang C, Wang QT, Liu H, Zhang ZZ, Huang WL. Advancement and prospects of tumor gene therapy. CHINESE JOURNAL OF CANCER 2011; 30:182-8. [PMID: 21352695 PMCID: PMC4013314 DOI: 10.5732/cjc.010.10074] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/20/2010] [Revised: 03/15/2010] [Accepted: 04/15/2010] [Indexed: 12/17/2022]
Abstract
Gene therapy is one of the most attractive fields in tumor therapy. In past decades, significant progress has been achieved. Various approaches, such as viral and non-viral vectors and physical methods, have been developed to make gene delivery safer and more efficient. Several therapeutic strategies have evolved, including gene-based (tumor suppressor genes, suicide genes, antiangiogenic genes, cytokine and oxidative stress-based genes) and RNA-based (antisense oligonucleotides and RNA interference) approaches. In addition, immune response-based strategies (dendritic cell- and T cell-based therapy) are also under investigation in tumor gene therapy. This review highlights the progress and recent developments in gene delivery systems, therapeutic strategies, and possible clinical directions for gene therapy.
Collapse
Affiliation(s)
- Chao Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, P. R. China;
| | - Qing-Tao Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, P. R. China;
| | - He Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, P. R. China;
| | - Zhen-Zhu Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, P. R. China;
| | - Wen-Lin Huang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, P. R. China;
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong 510060, P. R. China;
- Research Department, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P. R. China
| |
Collapse
|
28
|
Pajtasz-Piasecka E, Indrová M. Dendritic cell-based vaccines for the therapy of experimental tumors. Immunotherapy 2010; 2:257-68. [PMID: 20635932 DOI: 10.2217/imt.10.7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Dendritic cells (DCs) are believed to be the most potent antigen-presenting cells able to link the innate and adaptive immune systems. Many studies have focused on different immunotherapeutic approaches to applying DCs as tools to improve anticancer therapy. Although a number of investigations suggesting the benefit of DC-based vaccination during anticancer therapy have been reported, the general knowledge regarding the ultimate methods of DC-vaccine preparation is still unsatisfactory. In this article, the perspectives of DC-based anti-tumor immunotherapy and optimizing strategies of DC vaccination in humans in light of results obtained in mouse models are discussed.
Collapse
Affiliation(s)
- Elzbieta Pajtasz-Piasecka
- Ludwik Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland.
| | | |
Collapse
|
29
|
Lampen MH, Verweij MC, Querido B, van der Burg SH, Wiertz EJHJ, van Hall T. CD8+ T cell responses against TAP-inhibited cells are readily detected in the human population. THE JOURNAL OF IMMUNOLOGY 2010; 185:6508-17. [PMID: 20980626 DOI: 10.4049/jimmunol.1001774] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Target cell recognition by CTLs depends on the presentation of peptides by HLA class I molecules. Tumors and herpes viruses have adopted strategies to greatly hamper this peptide presentation at the important bottleneck, the peptide transporter TAP. Previously, we described the existence of a CD8(+) CTL subpopulation that selectively recognizes such TAP-deficient cells in mouse models. In this study, we show that the human counterpart of this CTL subset is readily detectable in healthy subjects. Autologous PBMC cultures were initiated with dendritic cells rendered TAP-impaired by gene transfer of the viral evasion molecule UL49.5. Strikingly, specific reactivity to B-LCLs expressing one of the other viral TAP-inhibitors (US6, ICP47, or BNLF2a) was already observed after three rounds of stimulation. These short-term T cell cultures and isolated CD8(+) CTL clones derived thereof did not recognize the normal B-LCL, indicating that the cognate peptide-epitopes emerge at the cell surface upon an inhibition in the MHC class I processing pathway. A diverse set of TCRs was used by the clones, and the cellular reactivity was TCR-dependent and HLA class I-restricted, implying the involvement of a broad antigenic peptide repertoire. Our data indicate that the human CD8(+) T cell pool comprises a diverse reactivity to target cells with impairments in the intracellular processing pathway, and these might be exploited for cancers that are associated with such defects and for infections with immune-evading herpes viruses.
Collapse
Affiliation(s)
- Margit H Lampen
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
30
|
Pan K, Zhao JJ, Wang H, Li JJ, Liang XT, Sun JC, Chen YB, Ma HQ, Liu Q, Xia JC. Comparative analysis of cytotoxic T lymphocyte response induced by dendritic cells loaded with hepatocellular carcinoma -derived RNA or cell lysate. Int J Biol Sci 2010; 6:639-48. [PMID: 20975822 PMCID: PMC2962266 DOI: 10.7150/ijbs.6.639] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2010] [Accepted: 08/29/2010] [Indexed: 02/02/2023] Open
Abstract
The choice of the tumor antigen preparation used for dendritic cell (DC) loading is important for optimizing DC vaccines. In the present study, we compared DCs pulsed with hepatocellular carcinoma (HCC) total RNA or cell lysates for their capacity to activate T cells. We showed here that HCC total RNA pulsed-DCs induced effector T lymphocyte responses which showed higher killing ability to HCC cell lines, as well as higher frequency of IFN-γ producing of CD4+ and CD8+ T cells when compared with lysate pulsed-DCs. Both of RNA and lysate loading did not influence the changes of mature DC phenotype and the capacity of inducing T cell proliferation. However, HCC lysate loading significantly inhibited the production of inflammatory cytokines IL-12p70, IFN-γ and enhanced the secretion of anti-inflammatory cytokines IL-10 of mature DCs. Our results indicated that DCs loaded with HCC RNA are superior to that loaded with lysate in priming anti-HCC CTL response, suggesting that total RNA may be a better choice for DCs-based HCC immunotherapy.
Collapse
Affiliation(s)
- Ke Pan
- State Key Laboratory of Oncology in Southern China and Department of Experimental Research, Cancer Center, Sun Yat-sen University, Guangzhou 510060, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Strategies for cancer vaccine development. J Biomed Biotechnol 2010; 2010. [PMID: 20706612 PMCID: PMC2914453 DOI: 10.1155/2010/596432] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2010] [Accepted: 05/17/2010] [Indexed: 12/29/2022] Open
Abstract
Treating cancer with vaccines has been a challenging field of investigation since the 1950s. Over the years, the lack of effective active immunotherapies has led to the development of numerous novel strategies. However, the use of therapeutic cancer vaccines may be on the verge of becoming an effective modality. Recent phase II/III clinical trials have achieved hopeful results in terms of overall survival. Yet despite these encouraging successes, in general, very little is known about the basic immunological mechanisms involved in vaccine immunotherapy. Gaining a better understanding of the mechanisms that govern the specific immune responses (i.e., cytotoxic T lymphocytes, CD4 T helper cells, T regulatory cells, cells of innate immunity, tumor escape mechanisms) elicited by each of the various vaccine platforms should be a concern of cancer vaccine clinical trials, along with clinical benefits. This review focuses on current strategies employed by recent clinical trials of therapeutic cancer vaccines and analyzes them both clinically and immunologically.
Collapse
|
32
|
Direct and indirect antitumor effects by human peripheral blood lymphocytes expressing both chimeric immune receptor and interleukin-2 in ovarian cancer xenograft model. Cancer Gene Ther 2010; 17:742-50. [PMID: 20559334 DOI: 10.1038/cgt.2010.30] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Human peripheral blood lymphocytes (PBLs) electroporated with RNA encoding anti-Her-2/neu-specific chimeric immune receptor (CIR) have been reported to elicit potent immune responses against SKOV3 tumors in a nude mouse model. However, CIR-electroporated PBL (CIR-PBL) did not proliferate, and the cell number rapidly decreased in the absence of exogenous interleukin-2 (IL-2). In this study, PBLs electroporated with both CIR and IL-2 RNA (CIR/IL-2-PBL) were studied to determine whether antitumor effects could be improved by adoptive immunotherapy. CIR and IL-2 were expressed in CIR/IL-2-PBL at levels similar to PBLs electroporated, with IL-2 RNA (IL-2-PBL) or CIR-PBL. Transfer of IL-2 RNA induced proliferation and prolonged survival of PBLs in vitro. In a xenograft model, both IL-2-PBL and CIR/IL-2-PBL showed significantly higher antitumor effects than CIR-PBL. The number of tumor-infiltrating natural killer (NK) cells was significantly increased in IL-2-PBL and CIR/IL-2-PBL. After NK cell depletion, IL-2-PBL showed significantly lower antitumor effects than CIR/IL-2-PBL. These results suggest that transfer of IL-2 RNA to CIR-PBL can promote NK cell infiltration of tumors and prolong survival of infused PBLs in vivo. RNA electroporated PBLs may represent efficient tools for delivery of functional molecules to tumors by multiple gene transfer.
Collapse
|
33
|
Herbert N, Haferkamp A, Schmitz-Winnenthal HF, Zöller M. Concomitant tumor and autoantigen vaccination supports renal cell carcinoma rejection. THE JOURNAL OF IMMUNOLOGY 2010; 185:902-16. [PMID: 20548033 DOI: 10.4049/jimmunol.0902683] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Efficient tumor vaccination frequently requires adjuvant. Concomitant induction of an autoimmune response is discussed as a means to strengthen a weak tumor Ag-specific response. We asked whether the efficacy of dendritic cell (DC) vaccination with the renal cell carcinoma Ags MAGE-A9 (MAGE9) and G250 could be strengthened by covaccination with the renal cell carcinoma autoantigen GOLGA4. BALB/c mice were vaccinated with DC loaded with MHC class I-binding peptides of MAGE9 or G250 or tumor lysate, which sufficed for rejection of low-dose RENCA-MAGE9 and RENCA-G250 tumor grafts, but only retarded tumor growth at 200 times the tumor dose at which 100% of animals will develop a tumor. Instead, 75-100% of mice prevaccinated concomitantly with Salmonella typhimurium transformed with GOLGA4 cDNA in a eukaryotic expression vector rejected 200 times the tumor dose at which 100% of animals will develop tumor. In a therapeutic setting, the survival rate increased from 20-40% by covaccination with S. typhimurium-GOLGA4. Autoantigen covaccination significantly strengthened tumor Ag-specific CD4(+) and CD8(+) T cell expansion, particularly in peptide-loaded DC-vaccinated mice. Covaccination was accompanied by an increase in inflammatory cytokines, boosted IL-12 and IFN-gamma expression, and promoted a high tumor Ag-specific CTL response. Concomitant autoantigen vaccination also supported CCR6, CXCR3, and CXCR4 upregulation and T cell recruitment into the tumor. It did not affect regulatory T cells, but slightly increased myeloid-derived suppressor cells. Thus, tumor cell eradication was efficiently strengthened by concomitant induction of an immune response against a tumor Ag and an autoantigen expressed by the tumor cell. Activation of autoantigen-specific Th cells strongly supports tumor-specific Th cells and thereby CTL activation.
Collapse
Affiliation(s)
- Nicolás Herbert
- Department of Tumor Cell Biology, University Hospital of Surgery, University of Heidelberg, Germany
| | | | | | | |
Collapse
|
34
|
Gholamin M, Moaven O, Farshchian M, Mahmoudi M, Sankian M, Memar B, Forghani MN, Malekzadeh R, Rajabi-Mashhadi MT, Abbaszadegan MR. Induction of cytotoxic T lymphocytes primed with tumor RNA-loaded dendritic cells in esophageal squamous cell carcinoma: preliminary step for DC vaccine design. BMC Cancer 2010; 10:261. [PMID: 20525404 PMCID: PMC2902443 DOI: 10.1186/1471-2407-10-261] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2009] [Accepted: 06/07/2010] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Dendritic cells (DC) are potent antigen presenting cells with the ability to prime naïve T cells and convert them to cytotoxic T-lymphocytes (CTL). We evaluated the capability of autologous DCs transfected with total tumor and normal RNA to induce cytotoxic CTL as the preliminary step to design a DC-based vaccine in the esophageal squamous cell carcinoma (ESCC). METHODS Monocytes-derived DCs were electroporated with either total tumor RNA or normal RNA. T cells were then primed with tumor RNA transfected DCs and lytic effects of the generated CTL were measured with Cytotoxicity assay and IFN-gamma Release Elispot assay. RESULTS Cytotoxicity was induced against DCs loaded with tumoral RNA (%24.8 +/- 5.2 SEM) while in normal RNA-loaded DCs, it was minimal (%6.1 +/- 2.4 SEM) and significantly lower (p < 0.05). INF-gamma secretion was more than 2-folds higher in tumoral RNA-loaded DCs when compared with normal RNA-loaded DCs (p < 0.05). CONCLUSION Electroporating DCs with tumor RNA generated tumor antigen presenting cells which in turn enhanced cytotoxic effects of the T cells against ESCC. This may be a useful autologous ex vivo screening tool for confirming the lytic effects of primed T cells on tumors and evaluate probable further adverse effects on noncancerous tissues. These data provide crucial preliminary information to establish a total tumor RNA-pulsed DC vaccine therapy of ESCC.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Cancer Vaccines/pharmacology
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/immunology
- Carcinoma, Squamous Cell/therapy
- Cell Separation
- Cells, Cultured
- Cytotoxicity Tests, Immunologic
- Cytotoxicity, Immunologic
- Dendritic Cells/immunology
- Dendritic Cells/transplantation
- Electroporation
- Esophageal Neoplasms/genetics
- Esophageal Neoplasms/immunology
- Esophageal Neoplasms/therapy
- Female
- Flow Cytometry
- Humans
- Immunophenotyping
- Interferon-gamma/immunology
- Lymphocyte Activation
- Male
- Middle Aged
- RNA, Messenger/immunology
- T-Lymphocytes, Cytotoxic/immunology
- Transfection
- Transplantation, Autologous
Collapse
Affiliation(s)
- Mehran Gholamin
- Division of Human Genetics, Immunology Research Center, Avicenna Research Institute, Mashhad University of Medical Sciences (MUMS), Mashhad, Iran
| | - Omeed Moaven
- Division of Human Genetics, Immunology Research Center, Avicenna Research Institute, Mashhad University of Medical Sciences (MUMS), Mashhad, Iran
| | - Moein Farshchian
- Division of Human Genetics, Immunology Research Center, Avicenna Research Institute, Mashhad University of Medical Sciences (MUMS), Mashhad, Iran
| | - Mahmoud Mahmoudi
- Department of Molecular Immunology, Immunology Research Center, MUMS, Mashhad, Iran
| | - Mojtaba Sankian
- Department of Immunobiochemistry, Immunology Research Center, MUMS, Mashhad, Iran
| | - Bahram Memar
- Department of Pathology, Omid Hospital, MUMS, Mashhad, Iran
| | | | - Reza Malekzadeh
- Digestive Disease Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mohammad Reza Abbaszadegan
- Division of Human Genetics, Immunology Research Center, Avicenna Research Institute, Mashhad University of Medical Sciences (MUMS), Mashhad, Iran
| |
Collapse
|
35
|
Kuhn AN, Diken M, Kreiter S, Selmi A, Kowalska J, Jemielity J, Darzynkiewicz E, Huber C, Türeci O, Sahin U. Phosphorothioate cap analogs increase stability and translational efficiency of RNA vaccines in immature dendritic cells and induce superior immune responses in vivo. Gene Ther 2010; 17:961-71. [PMID: 20410931 DOI: 10.1038/gt.2010.52] [Citation(s) in RCA: 171] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Vaccination with in vitro transcribed RNA coding for tumor antigens is considered a promising approach for cancer immunotherapy and has already entered human clinical testing. One of the basic objectives for development of RNA as a drug is the optimization of immunobioavailability of the encoded antigen in vivo. By analyzing the effect of different synthetic 5' mRNA cap analogs on the kinetics of the encoded protein, we found that m(2)(7,2'-O)Gpp(S)pG (beta-S-ARCA) phosphorothioate caps, in particular the D1 diastereoisomer, profoundly enhance RNA stability and translational efficiency in immature but not mature dendritic cells. Moreover, in vivo delivery of the antigen as beta-S-ARCA(D1)-capped RNA species is superior for protein expression and for efficient priming and expansion of naïve antigen-specific T cells in mice. Our findings establish 5' mRNA cap analogs as yet another module for tuning immunopharmacological properties of recombinant antigen-encoding RNA for vaccination purposes.
Collapse
Affiliation(s)
- A N Kuhn
- Division of Translational and Experimental Oncology, Department of Internal Medicine III, Johannes Gutenberg-University, Mainz, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Bøe S, Sæbøe-Larssen S, Hovig E. Light-Induced Gene Expression Using Messenger RNA Molecules. Oligonucleotides 2010; 20:1-6. [DOI: 10.1089/oli.2009.0209] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Sigurd Bøe
- Department of Tumor Biology, The Norwegian Radium Hospital, Rikshospitalet HF, Oslo, Norway
| | - Stein Sæbøe-Larssen
- Department of Immunology, Institute of Cancer Research, The Norwegian Radium Hospital, Rikshospitalet HF, Oslo, Norway
| | - Eivind Hovig
- Department of Tumor Biology, The Norwegian Radium Hospital, Rikshospitalet HF, Oslo, Norway
| |
Collapse
|
37
|
Abstract
Telomerase is highly expressed in essentially all cancer forms, while the expression in normal tissues is restricted. Moreover, telomerase activity is considered indispensable for tumor immortalization and growth. Human telomerase reverse transcriptase (hTERT), the rate-limiting subunit of the telomerase complex, is therefore an attractive target for cancer vaccination. The present review provides an update on the development of GV1001, a peptide vaccine representing a 16-aa hTERT sequence. GV1001 binds multiple HLA class II molecules and harbors putative HLA class I epitopes. The peptide may therefore elicit combined CD4/CD8 T-cell responses, considered important to initiate tumor eradication and long-term memory. Phase I/II trials in advanced pancreatic and pulmonary cancer patients have demonstrated GV1001-specific T-cell responses in > 50% of subjects, without clinically important toxicity. The results indicate a correlation between development of GV1001-specific responses and prolonged survival. However, as in most cancer vaccine trials, a large proportion of immune responders experience no clinical benefit. Long-term survivors harbor durable GV1001-specific T-cell responses with high IFN-gamma/IL-10 ratios and polyfunctional cytokine patterns. Interestingly, the cytokine profiles do not follow a T(H)1/T(H)2 delineation. Here, the author discusses how immunomonitoring may be improved to discriminate between efficient and pointless immune responses, and which questions to address in the further development of GV1001.
Collapse
Affiliation(s)
- Jon Amund Kyte
- Department of Clinical Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0310 Oslo, Norway. jon.amund.kyte@rr research.no
| |
Collapse
|
38
|
Abstract
Adoptive transfer of T cells with restricted tumor specificity provides a promising approach to immunotherapy of cancers. However, the isolation of autologous cytotoxic T cells that recognize tumor-associated antigens is time consuming and fails in many instances. Alternatively, gene modification with tumor antigen-specific T-cell receptors (TCR) or chimeric antigen receptors (CARs) can be used to redirect the specificity of large numbers of immune cells toward the malignant cells. Chimeric antigen receptors are composed of the single-chain variable fragment (scFv) of a tumor-recognizing antibody cloned in frame with human T-cell signaling domains (e.g., CD3zeta, CD28, OX40, 4-1BB), thus combining the specificity of antibodies with the effector functions of cytotoxic T cells. Upon antigen binding, the intracellular signaling domains of the CAR initiate cellular activation mechanisms including cytokine secretion and cytolysis of the antigen-positive target cell.In this chapter, we provide detailed protocols for large-scale ex vivo expansion of T cells and manufacturing of medium-scale batches of CAR-expressing T cells for translational research by mRNA electroporation. An anti-CD19 chimeric receptor for the targeting of leukemias and lymphomas was used as a model system. We are currently scaling up the protocols to adapt them to cGMP production of a large number of redirected T cells for clinical applications.
Collapse
Affiliation(s)
- Hilde Almåsbak
- Department of Immunology, Radiumhospitalet-Rikshospitalet, University Hospital, Oslo, Norway
| | | | | |
Collapse
|
39
|
Van Nuffel AMT, Corthals J, Neyns B, Heirman C, Thielemans K, Bonehill A. Immunotherapy of cancer with dendritic cells loaded with tumor antigens and activated through mRNA electroporation. Methods Mol Biol 2010; 629:405-52. [PMID: 20387165 DOI: 10.1007/978-1-60761-657-3_27] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Since decades, the main goal of tumor immunologists has been to increase the capacity of the immune system to mediate tumor regression. Considerable progress has been made in enhancing the efficacy of therapeutic anticancer vaccines. First, dendritic cells (DCs) have been identified as the key players in orchestrating primary immune responses. A better understanding of their biology and the development of procedures to generate vast amounts of DCs in vitro have accelerated the development of potent immunotherapeutic strategies for cancer. Second, tumor-associated antigens have been identified which are either selectively or preferentially expressed by tumor cells and can be recognized by the immune system. Finally, several studies have been performed on the genetic modification of DCs with tumor antigens. In this regard, loading the DCs with mRNA, which enables them to produce/process and present the tumor antigens themselves, has emerged as a promising strategy. Here, we will first overview the different aspects that must be taken into account when generating an mRNA-based DC vaccine and the published clinical studies exploiting mRNA-loaded DCs. Second, we will give a detailed description of a novel procedure to generate a vaccine consisting of tumor antigen-expressing dendritic cells with an in vitro superior capacity to induce anti-tumor immune responses. Here, immature DCs are electroporated with mRNAs encoding a tumor antigen, CD40 ligand (CD40L), CD70, and constitutively active (caTLR4) to generate mature antigen-presenting DCs.
Collapse
Affiliation(s)
- An M T Van Nuffel
- Laboratory of Molecular and Cellular Therapy, Department of Physiology - Immunology, Medical School of the Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | | | | | | | | | | |
Collapse
|
40
|
Smits EL, Anguille S, Cools N, Berneman ZN, Van Tendeloo VF. Dendritic Cell-Based Cancer Gene Therapy. Hum Gene Ther 2009; 20:1106-18. [DOI: 10.1089/hum.2009.145] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Evelien L.J.M. Smits
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, B-2610 Wilrijk (Antwerp), Belgium
| | - Sébastien Anguille
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, B-2610 Wilrijk (Antwerp), Belgium
- Center for Cell Therapy and Regenerative Medicine (CCTRM), Antwerp University Hospital, B-2650 Edegem (Antwerp), Belgium
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, B-2610 Wilrijk (Antwerp), Belgium
| | - Zwi N. Berneman
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, B-2610 Wilrijk (Antwerp), Belgium
- Center for Cell Therapy and Regenerative Medicine (CCTRM), Antwerp University Hospital, B-2650 Edegem (Antwerp), Belgium
| | - Viggo F.I. Van Tendeloo
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, B-2610 Wilrijk (Antwerp), Belgium
- Center for Cell Therapy and Regenerative Medicine (CCTRM), Antwerp University Hospital, B-2650 Edegem (Antwerp), Belgium
| |
Collapse
|
41
|
Cruz M, Enes M, Pereira M, Dourado M, Sarmento Ribeiro AB. [Experimental models in oncology: contribution of cell culture on understanding the biology of cancer]. REVISTA PORTUGUESA DE PNEUMOLOGIA 2009; 15:669-82. [PMID: 19547898 DOI: 10.1016/s0873-2159(15)30163-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
In the beginning of the 20th century, tissue culture was started with the aim of studying the behaviour of animal cells in normal and stress conditions. The cell study at molecular level depends on their capacity of growing and how they can be manipulated in laboratory. In vitro cell culture allows us the possibility of studying biological key processes, such as growth, differentiation and cell death, and also to do genetic manipulations essential to the knowledge of structure and genes function. Human stem cells culture provides strategies to circumvent other models' deficiencies. It seems that cancer stem cells remain quiescent until activation by appropriated micro-environmental stimulation. Several studies reveal that different cancer types could be due to stem cell malignant transformations. Removal of these cells is essential to the development of more effective cancer therapies for advanced disease. On the other hand, dendritic cells modified in culture may be used as a therapeutic vaccine in order to induce tumour withdraw.
Collapse
Affiliation(s)
- Mariana Cruz
- Faculdade de Medicina da Universidade de Coimbra, Coimbra, Portugal
| | | | | | | | | |
Collapse
|
42
|
Feyerabend S, Stevanovic S, Gouttefangeas C, Wernet D, Hennenlotter J, Bedke J, Dietz K, Pascolo S, Kuczyk M, Rammensee HG, Stenzl A. Novel multi-peptide vaccination in Hla-A2+ hormone sensitive patients with biochemical relapse of prostate cancer. Prostate 2009; 69:917-27. [PMID: 19267352 DOI: 10.1002/pros.20941] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND A phase I/II trial was conducted to assess feasibility and tolerability of tumor associated antigen peptide vaccination in hormone sensitive prostate carcinoma (PC) patients with biochemical recurrence after primary surgical treatment. METHODS Nineteen HLA-A2 positive patients with rising PSA without detectable metastatic disease or local recurrence received 11 HLA-A*0201-restricted and two HLA class II synthetic peptides derived from PC tumor antigens subcutaneously for 18 months or until PSA progression. The vaccine was emulgated in montanide ISA51 and combined with imiquimod, GM-CSF, mucin-1-mRNA/protamine complex, local hyperthermia or no adjuvant. PSA was assessed, geometric mean doubling times (DT) calculated and clinical performance monitored. RESULTS PSA DT of 4 out of 19 patients (21%) increased from 4.9 to 25.8 months during vaccination. Out of these, two patients (11%) exhibited PSA stability for 28 and 31 months which were still continuing at data cut-off. One patient showed no change of PSA DT during vaccination but decline after the therapy. Three patients had an interim PSA decline or DT increase followed by DT decrease compared to baseline PSA DT. Three of the responding patients received imiquimod and one the mucin-1-mRNA/protamine complex as adjuvant; both are Toll-like receptor-7 agonists. Eleven (58%) patients had progressive PSA values. The vaccine was well tolerated, and no grade III or IV toxicity occurred. CONCLUSION Multi-peptide vaccination stabilized or slowed down PSA progress in four of 19 cases. The vaccination approach is promising with moderate adverse events. Long-term stability delayed androgen deprivation up to 31 months. TLR-7 co-activation seems to be beneficial.
Collapse
|
43
|
Koh YT, Gray A, Higgins SA, Hubby B, Kast WM. Androgen ablation augments prostate cancer vaccine immunogenicity only when applied after immunization. Prostate 2009; 69:571-84. [PMID: 19143030 PMCID: PMC2732563 DOI: 10.1002/pros.20906] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Androgen ablation (AA) causes apoptosis of normal and neoplastic prostate cells. It is a standard treatment for advanced prostate cancer. Androgen ablation-mediated immunological effects include bone marrow hyperplasia, thymic regeneration, T and B cell lymphopoeisis and restoration of age-related peripheral T cell dysfunction. Androgens also regulate the transcription of several cytokines. Dendritic cells (DC) are the most potent antigen presenting cells that can activate antigen-specific naïve T cells. Despite myriad clinical trials involving DC-based prostate cancer immunotherapies, the effects of AA on DC function remain largely uncharacterized. Therefore, we investigated the effects of AA on DC and whether it could improve the efficacy of prostate cancer immunotherapy. METHODS Cytokine expression changes due to AA were quantified by multiplex ELISA. Flow cytometry was used to assess AA-mediated effects on DC maturation and expression of costimulatory markers. Mixed leukocyte reactions and cell-mediated lysis assays elucidated the role of androgens in DC function. The effect of AA on the efficacy of vaccination against a prostate tumor-associated antigen was tested using Elispot assays. RESULTS Androgen ablation increased dendritic cell maturation and costimulatory marker expression, but had no effect on DC costimulatory function. However, DC isolated from castrated mice increased the expression of key cytokines by antigen-experienced T cells while decreasing their expression in naïve cells. Finally, androgen ablation improved immune responses to vaccination only when applied after immunization. CONCLUSION Androgen ablation causes differential effects of DC on primary and secondary T cell responses, thus augmenting vaccine immunogenicity only when applied after immunization.
Collapse
Affiliation(s)
- Yi T. Koh
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033
- Department of Molecular Microbiology & Immunology, University of Southern California, Los Angeles, CA 90033
| | - Andrew Gray
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033
| | - Sean A. Higgins
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033
| | - Bolyn Hubby
- Alphavax Inc., Research Triangle Park, NC 27709
| | - W. Martin Kast
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033
- Department of Molecular Microbiology & Immunology, University of Southern California, Los Angeles, CA 90033
- Department of Obstetrics & Gynecology, University of Southern California, Los Angeles, CA 90033
- Correspondence: W. Martin Kast, PhD, Norris Comprehensive Cancer Center, NRT 7507, University of Southern California, 1450 Biggy Street, Los Angeles, CA 90033, Phone: 1 323 442 3870, E-mail:
| |
Collapse
|
44
|
Pham W, Kobukai S, Hotta C, Gore JC. Dendritic cells: therapy and imaging. Expert Opin Biol Ther 2009; 9:539-64. [DOI: 10.1517/14712590902867739] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Wellington Pham
- Vanderbilt University, Institute of Imaging Science, 1161 21st Avenue South, AA. 1105 MCN, Nashville, TN 37232-2310, USA
| | - Saho Kobukai
- Vanderbilt University, Institute of Imaging Science, 1161 21st Avenue South, AA. 1105 MCN, Nashville, TN 37232-2310, USA
- *These individuals contributed equally to this work
| | - Chie Hotta
- Brigham and Women's Hospital, Harvard Medical School, Center for Neurologic Diseases, 77 Avenue Louis Pasteur, HIM 780, Boston, MA 02115, USA
- *These individuals contributed equally to this work
| | - John C Gore
- Vanderbilt University, Institute of Imaging Science, 1161 21st Avenue South, AA. 1105 MCN, Nashville, TN 37232-2310, USA
| |
Collapse
|
45
|
Simon T, Fonteneau JF, Grégoire M. Dendritic cell preparation for immunotherapeutic interventions. Immunotherapy 2009; 1:289-302. [DOI: 10.2217/1750743x.1.2.289] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Much effort has been made over the last decade to use dendritic cells (DCs) in vaccines to induce specific antitumor immune responses. However, the great hope provided by in vitro and in vivo preclinical investigations was not translated to the clinic in terms of clinical efficacy. Thus, one of the challenges resides in optimizing DC-based therapy to give maximum clinical efficacy while using manufacturing processes that enable quality control and scale-up of consistent products. In this article, we review DC biology and the DC-based clinical trials performed to date and focus on the DC maturation status compatible with the goals of cancer immunotherapy. We also highlight the different approaches used in these clinical studies, such as the DC types or subtypes used and their preparation. Finally, we discuss the immunological and clinical outcomes in treated patients, with emphasis on the strategies that could be used to improve DC-based vaccination.
Collapse
Affiliation(s)
- Thomas Simon
- INSERM U892, Institut de Biologie, 9 quai Moncousu, 44093 Nantes Cedex 01, France
| | | | - Marc Grégoire
- INSERM U892, Institut de Biologie, 9 quai Moncousu, 44093 Nantes Cedex 01, France
| |
Collapse
|
46
|
Birkholz K, Hombach A, Krug C, Reuter S, Kershaw M, Kämpgen E, Schuler G, Abken H, Schaft N, Dörrie J. Transfer of mRNA encoding recombinant immunoreceptors reprograms CD4+ and CD8+ T cells for use in the adoptive immunotherapy of cancer. Gene Ther 2009; 16:596-604. [DOI: 10.1038/gt.2008.189] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
47
|
Engell-Noerregaard L, Hansen TH, Andersen MH, thor Straten P, Svane IM. Review of clinical studies on dendritic cell-based vaccination of patients with malignant melanoma: assessment of correlation between clinical response and vaccine parameters. Cancer Immunol Immunother 2009; 58:1-14. [PMID: 18719915 PMCID: PMC11030652 DOI: 10.1007/s00262-008-0568-4] [Citation(s) in RCA: 146] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2008] [Accepted: 07/16/2008] [Indexed: 02/07/2023]
Abstract
During the past years numerous clinical trials have been carried out to assess the ability of dendritic cell (DC) based immunotherapy to induce clinically relevant immune responses in patients with malignant diseases. A broad range of cancer types have been targeted including malignant melanoma which in the disseminated stage have a very poor prognosis and only limited treatment options with moderate effectiveness. Herein we describe the results of a focused search of recently published clinical studies on dendritic cell vaccination in melanoma and review different vaccine parameters which are frequently claimed to have a possible influence on clinical response. These parameters include performance status, type of antigen, DC maturation status, route of vaccine administration, use of adjuvant, and vaccine induced immune response. In total, 38 articles found through Medline search, have been included for analysis covering a total of 626 patients with malignant melanoma treated with DC based therapy. Clinical response (CR, PR and SD) were found to be significantly correlated with the use of peptide antigens (p = 0.03), the use of any helper antigen/adjuvant (p = 0.002), and induction of antigen specific T cells (p = 0.0004). No significant correlations between objective response (CR and PR) and the tested parameters were found. However, a few non-significant trends were demonstrated; these included an association between objective response and use of immature DCs (p = 0.08), use of adjuvant (p = 0.09), and use of autologous antigen preparation (p = 0.12). The categorisation of SD in the response group is debatable. Nevertheless, when the SD group were analysed separately we found that SD was significantly associated with use of peptide antigens (p = 0.0004), use of adjuvant (p = 0.01), and induction of antigen specific T cells (p = 0.0003). No specific route of vaccine administration showed superiority. Important lessons can be learned from previous studies, interpretation of these findings should, however, be done with reservation for the many minor deviations in the different treatment schedules among the published studies, which were not considered in order to be able to process and group the data.
Collapse
Affiliation(s)
- Lotte Engell-Noerregaard
- Department of Oncology, Herlev Hospital, Herlev, Denmark
- Department of Hematology, Center for Cancer Immune Therapy (CCIT), Herlev Hospital, Herlev, Denmark
| | - Troels Holz Hansen
- Department of Hematology, Center for Cancer Immune Therapy (CCIT), Herlev Hospital, Herlev, Denmark
| | - Mads Hald Andersen
- Department of Hematology, Center for Cancer Immune Therapy (CCIT), Herlev Hospital, Herlev, Denmark
| | - Per thor Straten
- Department of Hematology, Center for Cancer Immune Therapy (CCIT), Herlev Hospital, Herlev, Denmark
| | - Inge Marie Svane
- Department of Oncology, Herlev Hospital, Herlev, Denmark
- Department of Hematology, Center for Cancer Immune Therapy (CCIT), Herlev Hospital, Herlev, Denmark
| |
Collapse
|
48
|
Abstract
Dendritic cells (DC) have profound abilities to induce and coordinate T-cell immunity. This makes them ideal biological agents for use in immunotherapeutic strategies to augment T-cell immunity to HIV infection. Current clinical trials are administering DC-HIV antigen preparations carried out ex vivo as proof of principle that DC immunotherapy is safe and efficacious in HIV-infected patients. These trials are largely dependent on preclinical studies that will provide knowledge and guidance about the types of DC, form of HIV antigen, method of DC maturation, route of DC administration, measures of anti-HIV immune function and ultimately control of HIV replication. Additionally, promising immunotherapy approaches are being developed based on targeting of DC with HIV antigens in vivo. The objective is to define a safe and effective strategy for enhancing control of HIV infection in patients undergoing antiretroviral therapy.
Collapse
Affiliation(s)
- C R Rinaldo
- Department of Infectious Diseases, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
49
|
Mikszta JA, Laurent PE. Cutaneous delivery of prophylactic and therapeutic vaccines: historical perspective and future outlook. Expert Rev Vaccines 2008; 7:1329-39. [PMID: 18980537 DOI: 10.1586/14760584.7.9.1329] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The skin has long been recognized as an attractive target for vaccine administration. A number of clinical studies have tested the epidermal and dermal routes of delivery using a variety of vaccines over the years. In many cases, cutaneous administration has been associated with immunological benefits, such as the induction of greater immune responses compared with those elicited by conventional routes of delivery. Furthermore, there is a growing body of evidence to suggest that such benefits may be particularly important for certain higher-risk populations, such as the elderly, the immunocompromised and cancer patients. Despite the potential advantages of vaccination via the skin, results have sometimes been conflicting and the full benefits of this approach have not been fully realized, partly due to the lack of delivery devices that accurately and reproducibly administer vaccines to the skin. The 5-year outlook, however, appears quite promising as new cutaneous delivery systems advance through clinical trials and become available for more widespread clinical and commercial use.
Collapse
Affiliation(s)
- John A Mikszta
- BD Technologies, 21 Davis Drive, Research Triangle Park, NC 27709, USA.
| | | |
Collapse
|
50
|
Narita M, Tochiki N, Saitoh A, Watanabe N, Kaji M, Satoh N, Yamahira A, Nakamura T, Masuko M, Furukawa T, Toba K, Fuse I, Aizawa Y, Takahashi M. Induction of antigen-specific cytotoxic T lymphocytes by using monocyte-derived DCs transfected with in vitro-transcribed WT1 or SART1 mRNA. Med Oncol 2008; 26:429-36. [DOI: 10.1007/s12032-008-9142-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2008] [Accepted: 11/20/2008] [Indexed: 10/21/2022]
|