1
|
Chhabra L, Pandey RK, Kumar R, Sundar S, Mehrotra S. Navigating the Roadblocks: Progress and Challenges in Cell-Based Therapies for Human Immunodeficiency Virus. J Cell Biochem 2024:e30669. [PMID: 39485037 DOI: 10.1002/jcb.30669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/26/2024] [Accepted: 10/11/2024] [Indexed: 11/03/2024]
Abstract
Cell-based therapies represent a major advancement in the treatment and management of HIV/AIDS, with a goal to overcome the limitations of traditional antiretroviral therapy (ART). These innovative approaches not only promise a functional cure by reconstructing the immune landscape but also address the persistent viral reservoirs. For example, stem cell therapies have emerged from the foundational success of allogeneic hematopoietic stem cell transplantation in curing HIV infection in a limited number of cases. B cell therapies make use of genetically modified B cells constitutively expressing broadly neutralizing antibodies (bNAbs) against target viral particles and infected cells. Adoptive cell transfer (ACT), including TCR-T therapy, CAR-T cells, NK-CAR cells, and DC-based therapy, is adapted from cancer immunotherapy and repurposed for HIV eradication. In this review, we summarize the mechanisms through which these engineered cells recognize and destroy HIV-infected cells, the modification strategies, and their role in sustaining remission in the absence of ART. The review also addresses the challenges to cell-based therapies against HIV and discusses the recent advancements aimed at overcoming them.
Collapse
Affiliation(s)
- Lakshay Chhabra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | | | - Rajiv Kumar
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Shyam Sundar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| |
Collapse
|
2
|
Toner K, McCann CD, Bollard CM. Applications of cell therapy in the treatment of virus-associated cancers. Nat Rev Clin Oncol 2024; 21:709-724. [PMID: 39160243 DOI: 10.1038/s41571-024-00930-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2024] [Indexed: 08/21/2024]
Abstract
A diverse range of viruses have well-established roles as the primary driver of oncogenesis in various haematological malignancies and solid tumours. Indeed, estimates suggest that approximately 1.5 million patients annually are diagnosed with virus-related cancers. The predominant human oncoviruses include Epstein-Barr virus (EBV), Kaposi sarcoma-associated herpesvirus (KSHV), hepatitis B and C viruses (HBV and HCV), human papillomavirus (HPV), human T-lymphotropic virus type 1 (HTLV1), and Merkel cell polyomavirus (MCPyV). In addition, although not inherently oncogenic, human immunodeficiency virus (HIV) is associated with immunosuppression that contributes to the development of AIDS-defining cancers (specifically, Kaposi sarcoma, aggressive B cell non-Hodgkin lymphoma and cervical cancer). Given that an adaptive T cell-mediated immune response is crucial for the control of viral infections, increasing research is being focused on evaluating virus-specific T cell therapies for the treatment of virus-associated cancers. In this Review, we briefly outline the roles of viruses in the pathogenesis of these malignancies before describing progress to date in the field of virus-specific T cell therapy and evaluating the potential utility of these therapies to treat or possibly even prevent virus-related malignancies.
Collapse
Affiliation(s)
- Keri Toner
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA
- Department of Paediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Chase D McCann
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA
- Department of Paediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Catherine M Bollard
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA.
- Department of Paediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA.
| |
Collapse
|
3
|
Parhiz H, Atochina-Vasserman EN, Weissman D. mRNA-based therapeutics: looking beyond COVID-19 vaccines. Lancet 2024; 403:1192-1204. [PMID: 38461842 DOI: 10.1016/s0140-6736(23)02444-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 07/06/2023] [Accepted: 10/30/2023] [Indexed: 03/12/2024]
Abstract
Recent advances in mRNA technology and its delivery have enabled mRNA-based therapeutics to enter a new era in medicine. The rapid, potent, and transient nature of mRNA-encoded proteins, without the need to enter the nucleus or the risk of genomic integration, makes them desirable tools for treatment of a range of diseases, from infectious diseases to cancer and monogenic disorders. The rapid pace and ease of mass-scale manufacturability of mRNA-based therapeutics supported the global response to the COVID-19 pandemic. Nonetheless, challenges remain with regards to mRNA stability, duration of expression, delivery efficiency, and targetability, to broaden the applicability of mRNA therapeutics beyond COVID-19 vaccines. By learning from the rapidly expanding preclinical and clinical studies, we can optimise the mRNA platform to meet the clinical needs of each disease. Here, we will summarise the recent advances in mRNA technology; its use in vaccines, immunotherapeutics, protein replacement therapy, and genomic editing; and its delivery to desired specific cell types and organs for development of a new generation of targeted mRNA-based therapeutics.
Collapse
Affiliation(s)
- Hamideh Parhiz
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Drew Weissman
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
4
|
Zhang X, Li Y, Zhou Z. Lipid Nanoparticle-Based Delivery System-A Competing Place for mRNA Vaccines. ACS OMEGA 2024; 9:6219-6234. [PMID: 38371811 PMCID: PMC10870384 DOI: 10.1021/acsomega.3c08353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/24/2023] [Accepted: 12/29/2023] [Indexed: 02/20/2024]
Abstract
mRNA, as one of the foci of biomedical research in the past decade, has become a candidate vaccine solution for various infectious diseases and tumors and for regenerative medicine and immunotherapy due to its high efficiency, safety, and effectiveness. A stable and effective delivery system is needed to protect mRNAs from nuclease degradation while also enhancing immunogenicity. The success of mRNA lipid nanoparticles in treating COVID-19, to a certain extent, marks a milestone for mRNA vaccines and also promotes further research on mRNA delivery systems. Here, we explore mRNA vaccine delivery systems, especially lipid nanoparticles (LNPs), considering the current research status, prospects, and challenges of lipid nanoparticles, and explore other mRNA delivery systems.
Collapse
Affiliation(s)
- Xinyu Zhang
- Research
Center for Infectious Diseases, Tianjin
University of Traditional Chinese Medicine, 300193 Tianjin, China
- Institute
for Biological Product Control, National
Institutes for Food and Drug Control (NIFDC) and WHO Collaborating
Center for Standardization and Evaluation of Biologicals, No.31 Huatuo Street, Daxing District, 102629 Beijing, China
- College
of Life Science, Jilin University, 130012 Changchun, China
| | - Yuanfang Li
- Department
of Neurology, Zhongshan Hospital (Xiamen Branch), Fudan University, 361015 Xiamen, Fujian China
| | - Zehua Zhou
- Research
Center for Infectious Diseases, Tianjin
University of Traditional Chinese Medicine, 300193 Tianjin, China
| |
Collapse
|
5
|
Wang AYL, Chang YC, Chen KH, Loh CYY. Potential Application of Modified mRNA in Cardiac Regeneration. Cell Transplant 2024; 33:9636897241248956. [PMID: 38715279 PMCID: PMC11080755 DOI: 10.1177/09636897241248956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 03/26/2024] [Accepted: 04/07/2024] [Indexed: 05/12/2024] Open
Abstract
Heart failure remains the leading cause of human death worldwide. After a heart attack, the formation of scar tissue due to the massive death of cardiomyocytes leads to heart failure and sudden death in most cases. In addition, the regenerative ability of the adult heart is limited after injury, partly due to cell-cycle arrest in cardiomyocytes. In the current post-COVID-19 era, urgently authorized modified mRNA (modRNA) vaccines have been widely used to prevent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Therefore, modRNA-based protein replacement may act as an alternative strategy for improving heart disease. It is a safe, effective, transient, low-immunogenic, and integration-free strategy for in vivo protein expression, in addition to recombinant protein and stem-cell regenerative therapies. In this review, we provide a summary of various cardiac factors that have been utilized with the modRNA method to enhance cardiovascular regeneration, cardiomyocyte proliferation, fibrosis inhibition, and apoptosis inhibition. We further discuss other cardiac factors, modRNA delivery methods, and injection methods using the modRNA approach to explore their application potential in heart disease. Factors for promoting cardiomyocyte proliferation such as a cocktail of three genes comprising FoxM1, Id1, and Jnk3-shRNA (FIJs), gp130, and melatonin have potential to be applied in the modRNA approach. We also discuss the current challenges with respect to modRNA-based cardiac regenerative medicine that need to be overcome to apply this approach to heart disease. This review provides a short description for investigators interested in the development of alternative cardiac regenerative medicines using the modRNA platform.
Collapse
Affiliation(s)
- Aline Yen Ling Wang
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yun-Ching Chang
- Department of Health Industry Technology Management, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Kuan-Hung Chen
- Department of Physical Medicine & Rehabilitation, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | | |
Collapse
|
6
|
Sauerer T, Albrecht L, Sievers NM, Gerer KF, Hoyer S, Dörrie J, Schaft N. Electroporation of mRNA as a Universal Technology Platform to Transfect a Variety of Primary Cells with Antigens and Functional Proteins. Methods Mol Biol 2024; 2786:219-235. [PMID: 38814397 DOI: 10.1007/978-1-0716-3770-8_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Electroporation (EP) of mRNA into human cells is a broadly applicable method to transiently express proteins of choice in a variety of different cell types. We have spent more than two decades to optimize and adapt this method, first for antigen-loading of dendritic cells (DCs) and subsequently for T cells, B cells, bulk PBMCs, and several cell lines. In this regard, antigens were introduced, processed, and presented in context of MHC class I and II. Next to that, functional proteins like adhesion receptors, T-cell receptors (TCRs), chimeric antigen receptors (CARs), constitutively active signal transducers (i.e. caIKK), and others were successfully expressed. We have also established this protocol under full GMP compliance as part of a manufacturing license to produce mRNA-electroporated DCs and mRNA-electroporated T cells for therapeutic applications in clinical trials. Therefore, we here want to share our universal mRNA electroporation protocol and the experience we have gathered with this method. The advantages of the transfection method presented here are: (1) easy adaptation to different cell types; (2) scalability from 106 to approximately 108 cells per shot; (3) high transfection efficiency (80-99%); (4) homogenous protein expression; (5) GMP compliance if the EP is performed in a class A clean room; and (6) no transgene integration into the genome. The provided protocol involves: OptiMEM® as EP medium, a square-wave pulse with 500 V, and 4 mm cuvettes. To adapt the protocol to differently sized cells, simply the pulse time has to be altered. Thus, we share an overview of proven electroporation settings (including recovery media), which we have established for various cell types. Next to the basic protocol, we also provide an extensive list of hints and tricks, which, in our opinion, are of great value for everyone who intends to use this transfection technique.
Collapse
Affiliation(s)
- Tatjana Sauerer
- RNA-based Immunotherapy, Department of Dermatology, Universitätsklinikum Erlangen (UKER), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Leoni Albrecht
- RNA-based Immunotherapy, Department of Dermatology, Universitätsklinikum Erlangen (UKER), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Nico M Sievers
- RNA-based Immunotherapy, Department of Dermatology, Universitätsklinikum Erlangen (UKER), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Kerstin F Gerer
- RNA-based Immunotherapy, Department of Dermatology, Universitätsklinikum Erlangen (UKER), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
- Novartis Pharma GmbH, Nuremberg, Germany
| | - Stefanie Hoyer
- RNA-based Immunotherapy, Department of Dermatology, Universitätsklinikum Erlangen (UKER), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
- Department of Palliative Medicine, Universitätsklinikum Erlangen, Comprehensive Cancer Center CCC Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Jan Dörrie
- RNA-based Immunotherapy, Department of Dermatology, Universitätsklinikum Erlangen (UKER), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Niels Schaft
- RNA-based Immunotherapy, Department of Dermatology, Universitätsklinikum Erlangen (UKER), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany.
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany.
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany.
| |
Collapse
|
7
|
Banoun H. mRNA: Vaccine or Gene Therapy? The Safety Regulatory Issues. Int J Mol Sci 2023; 24:10514. [PMID: 37445690 DOI: 10.3390/ijms241310514] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
COVID-19 vaccines were developed and approved rapidly in response to the urgency created by the pandemic. No specific regulations existed at the time they were marketed. The regulatory agencies therefore adapted them as a matter of urgency. Now that the pandemic emergency has passed, it is time to consider the safety issues associated with this rapid approval. The mode of action of COVID-19 mRNA vaccines should classify them as gene therapy products (GTPs), but they have been excluded by regulatory agencies. Some of the tests they have undergone as vaccines have produced non-compliant results in terms of purity, quality and batch homogeneity. The wide and persistent biodistribution of mRNAs and their protein products, incompletely studied due to their classification as vaccines, raises safety issues. Post-marketing studies have shown that mRNA passes into breast milk and could have adverse effects on breast-fed babies. Long-term expression, integration into the genome, transmission to the germline, passage into sperm, embryo/fetal and perinatal toxicity, genotoxicity and tumorigenicity should be studied in light of the adverse events reported in pharmacovigilance databases. The potential horizontal transmission (i.e., shedding) should also have been assessed. In-depth vaccinovigilance should be carried out. We would expect these controls to be required for future mRNA vaccines developed outside the context of a pandemic.
Collapse
|
8
|
Laeremans T, den Roover S, Lungu C, D’haese S, Gruters RA, Allard SD, Aerts JL. Autologous dendritic cell vaccination against HIV-1 induces changes in natural killer cell phenotype and functionality. NPJ Vaccines 2023; 8:29. [PMID: 36864042 PMCID: PMC9980861 DOI: 10.1038/s41541-023-00631-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 02/20/2023] [Indexed: 03/04/2023] Open
Abstract
Although natural killer (NK) cells have been studied in connection with dendritic cell (DC)-based vaccination in the field of cancer immunology, their role has barely been addressed in the context of therapeutic vaccination against HIV-1. In this study, we evaluated whether a therapeutic DC-based vaccine consisting of monocyte-derived DCs electroporated with Tat, Rev and Nef encoding mRNA affects NK cell frequency, phenotype and functionality in HIV-1-infected individuals. Although the frequency of total NK cells did not change, we observed a significant increase in cytotoxic NK cells following immunisation. In addition, significant changes in the NK cell phenotype associated with migration and exhaustion were observed together with increased NK cell-mediated killing and (poly)functionality. Our results show that DC-based vaccination has profound effects on NK cells, which highlights the importance of evaluating NK cells in future clinical trials looking at DC-based immunotherapy in the context of HIV-1 infection.
Collapse
Affiliation(s)
- Thessa Laeremans
- grid.8767.e0000 0001 2290 8069Neuro-Aging and Viro-Immunotherapy Research Group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sabine den Roover
- grid.8767.e0000 0001 2290 8069Neuro-Aging and Viro-Immunotherapy Research Group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Cynthia Lungu
- grid.5645.2000000040459992XDepartment of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Sigrid D’haese
- grid.8767.e0000 0001 2290 8069Neuro-Aging and Viro-Immunotherapy Research Group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Rob A. Gruters
- grid.5645.2000000040459992XDepartment of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Sabine D. Allard
- grid.411326.30000 0004 0626 3362Department of Internal Medicine and Infectious Diseases, Universitair Ziekenhuis Brussel and Vrije Universiteit Brussel, Brussels, Belgium
| | - Joeri L. Aerts
- grid.8767.e0000 0001 2290 8069Neuro-Aging and Viro-Immunotherapy Research Group, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
9
|
Schroeder SM, Nelde A, Walz JS. Viral T-cell epitopes - Identification, characterization and clinical application. Semin Immunol 2023; 66:101725. [PMID: 36706520 DOI: 10.1016/j.smim.2023.101725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 01/26/2023]
Abstract
T-cell immunity, mediated by CD4+ and CD8+ T cells, represents a cornerstone in the control of viral infections. Virus-derived T-cell epitopes are represented by human leukocyte antigen (HLA)-presented viral peptides on the surface of virus-infected cells. They are the prerequisite for the recognition of infected cells by T cells. Knowledge of viral T-cell epitopes provides on the one hand a diagnostic tool to decipher protective T-cell immune responses in the human population and on the other hand various prophylactic and therapeutic options including vaccination approaches and the transfer of virus-specific T cells. Such approaches have already been proven to be effective against various viral infections, particularly in immunocompromised patients lacking sufficient humoral, antibody-based immune response. This review provides an overview on the state of the art as well as current studies regarding the identification and characterization of viral T-cell epitopes and approaches of clinical application. In the first chapter in silico prediction tools and direct, mass spectrometry-based identification of viral T-cell epitopes is compared. The second chapter provides an overview of commonly used assays for further characterization of T-cell responses and phenotypes. The final chapter presents an overview of clinical application of viral T-cell epitopes with a focus on human immunodeficiency virus (HIV), human cytomegalovirus (HCMV) and severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), being representatives of relevant viruses.
Collapse
Affiliation(s)
- Sarah M Schroeder
- Department of Peptide-based Immunotherapy, University and University Hospital Tübingen, Tübingen, Germany; Department for Otorhinolaryngology, Head, and Neck Surgery, University Hospital Tübingen, Tübingen, Germany; Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Annika Nelde
- Department of Peptide-based Immunotherapy, University and University Hospital Tübingen, Tübingen, Germany; Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany; Cluster of Excellence iFIT (EXC2180) 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
| | - Juliane S Walz
- Department of Peptide-based Immunotherapy, University and University Hospital Tübingen, Tübingen, Germany; Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany; Cluster of Excellence iFIT (EXC2180) 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany; Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany.
| |
Collapse
|
10
|
Cagigi A, Douradinha B. Have mRNA vaccines sentenced DNA vaccines to death? Expert Rev Vaccines 2023; 22:1154-1167. [PMID: 37941101 DOI: 10.1080/14760584.2023.2282065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 11/07/2023] [Indexed: 11/10/2023]
Abstract
INTRODUCTION After receiving emergency approval during the COVID-19 pandemic, mRNA vaccines have taken center stage in the quest to enhance future vaccination strategies for both infectious diseases and cancer. Indeed, they have significantly overshadowed another facet of genetic vaccination, specifically DNA vaccines. Nevertheless, it is important to acknowledge that both types of genetic vaccines have distinct advantages and disadvantages that set them apart from each other. AREAS COVERED In this work, we delve extensively into the history of genetic vaccines, their mechanisms of action, their strengths, and limitations, and ultimately highlight ongoing research in key areas for potential enhancement of both DNA and mRNA vaccines. EXPERT OPINION Here, we assess the significance of the primary benefits and drawbacks associated with DNA and mRNA vaccination. We challenge the current lines of thought by highlighting that the existing drawbacks of DNA vaccination could potentially be more straightforward to address compared to those linked with mRNA vaccination. In our view, this suggests that DNA vaccines should remain viable contenders in the pursuit of the future of vaccination.
Collapse
Affiliation(s)
- Alberto Cagigi
- Nykode Therapeutics ASA, Oslo Science Park, Oslo, Norway
| | | |
Collapse
|
11
|
Gene Therapy and Cardiovascular Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1396:235-254. [DOI: 10.1007/978-981-19-5642-3_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
|
12
|
Different dendritic cells-based vaccine constructs influence HIV-1 antigen-specific immunological responses and cytokine generation in virion-exposed splenocytes. Int Immunopharmacol 2022; 113:109406. [DOI: 10.1016/j.intimp.2022.109406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 10/18/2022] [Accepted: 10/28/2022] [Indexed: 11/09/2022]
|
13
|
Hertler J, Slama K, Schober B, Özrendeci Z, Marchand V, Motorin Y, Helm M. Synthesis of point-modified mRNA. Nucleic Acids Res 2022; 50:e115. [PMID: 36062567 PMCID: PMC9723659 DOI: 10.1093/nar/gkac719] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 07/20/2022] [Accepted: 08/15/2022] [Indexed: 12/24/2022] Open
Abstract
Synthetic mRNA has recently moved into the focus of therapeutic and vaccination efforts. Incorporation of modified nucleotides during in vitro transcription can improve translation and attenuate immunogenicity, but is limited to triphosphate nucleotides which are accepted by RNA polymerases, and their incorporation is either random or complete. In contrast, site-specific modification, herein termed 'point modification' in analogy to point mutations, holds significant technical challenge. We developed fundamental techniques for isolation of long, translatable and internally point-modified mRNAs. Enabling concepts include three-way-one-pot splint ligations, and isolation of mRNA by real-time elution from agarose gels. The use of blue light permitted visualization of mRNA in pre-stained gels without the photochemical damage associated with the use of hard UV-radiation. This allowed visualization of the mRNA through its migration in the agarose gel, which in turn, was a prerequisite for its recovery by electroelution into precast troughs. Co-eluting agarose particles were quantified and found to not be detrimental to mRNA translation in vitro. Translation of EGFP-coding mRNA into functional protein was quantified by incorporation of 35S-labelled methionine and by in-gel EGFP fluorescence. This enabled the functional analysis of point modifications, specifically of ribose methylations in the middle of a 1371 nt long mRNA.
Collapse
Affiliation(s)
- Jasmin Hertler
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-Universität, Staudinger Weg 5, D-55128 Mainz, Germany
| | - Kaouthar Slama
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-Universität, Staudinger Weg 5, D-55128 Mainz, Germany
| | - Benedikt Schober
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-Universität, Staudinger Weg 5, D-55128 Mainz, Germany
| | - Zeynep Özrendeci
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-Universität, Staudinger Weg 5, D-55128 Mainz, Germany
| | - Virginie Marchand
- IMoPA UMR7365 CNRS-UL, BioPole Université de Lorraine, Vandœuvre-lès-Nancy, France
| | - Yuri Motorin
- IMoPA UMR7365 CNRS-UL, BioPole Université de Lorraine, Vandœuvre-lès-Nancy, France
- Epitranscriptomics and RNA Sequencing (EpiRNA-Seq) Core Facility, UMS2008 IBSLor (CNRS-UL)/US40 (INSERM), Université de Lorraine, Vandœuvre-lès-Nancy, France
| | - Mark Helm
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-Universität, Staudinger Weg 5, D-55128 Mainz, Germany
| |
Collapse
|
14
|
mRNA Vaccines: Past, Present, Future. Asian J Pharm Sci 2022; 17:491-522. [PMID: 36105317 PMCID: PMC9459002 DOI: 10.1016/j.ajps.2022.05.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 05/11/2022] [Accepted: 05/23/2022] [Indexed: 11/23/2022] Open
Abstract
mRNA vaccines have emerged as promising alternative platforms to conventional vaccines. Their ease of production, low cost, safety profile and high potency render them ideal candidates for prevention and treatment of infectious diseases, especially in the midst of pandemics. The challenges that face in vitro transcribed RNA were partially amended by addition of tethered adjuvants or co-delivery of naked mRNA with an adjuvant-tethered RNA. However, it wasn't until recently that the progress made in nanotechnology helped enhance mRNA stability and delivery by entrapment in novel delivery systems of which, lipid nanoparticles. The continuous advancement in the fields of nanotechnology and tissue engineering provided novel carriers for mRNA vaccines such as polymeric nanoparticles and scaffolds. Various studies have shown the advantages of adopting mRNA vaccines for viral diseases and cancer in animal and human studies. Self-amplifying mRNA is considered today the next generation of mRNA vaccines and current studies reveal promising outcomes. This review provides a comprehensive overview of mRNA vaccines used in past and present studies, and discusses future directions and challenges in advancing this vaccine platform to widespread clinical use.
Collapse
|
15
|
de Almeida Baptista MV, da Silva LT, Samer S, Oshiro TM, Shytaj IL, Giron LB, Pena NM, Cruz N, Gosuen GC, Ferreira PRA, Cunha-Neto E, Galinskas J, Dias D, Sucupira MCA, de Almeida-Neto C, Salomão R, da Silva Duarte AJ, Janini LM, Hunter JR, Savarino A, Juliano MA, Diaz RS. Immunogenicity of personalized dendritic-cell therapy in HIV-1 infected individuals under suppressive antiretroviral treatment: interim analysis from a phase II clinical trial. AIDS Res Ther 2022; 19:2. [PMID: 35022035 PMCID: PMC8753935 DOI: 10.1186/s12981-021-00426-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/16/2021] [Indexed: 11/12/2022] Open
Abstract
Background We developed a personalized Monocyte-Derived Dendritic-cell Therapy (MDDCT) for HIV-infected individuals on suppressive antiretroviral treatment and evaluated HIV-specific T-cell responses. Methods PBMCs were obtained from 10 HIV+ individuals enrolled in trial NCT02961829. Monocytes were differentiated into DCs using IFN-α and GM-CSF. After sequencing each patient’s HIV-1 Gag and determining HLA profiles, autologous Gag peptides were selected based on the predicted individual immunogenicity and used to pulse MDDCs. Three doses of the MDDCT were administered every 15 days. To assess immunogenicity, patients’ cells were stimulated in vitro with autologous peptides, and intracellular IL-2, TNF, and interferon-gamma (IFN-γ) production were measured in CD4+ and CD8+ T-cells. Results The protocol of ex-vivo treatment with IFN-α and GM-CSF was able to induce maturation of MDDCs, as well as to preserve their viability for reinfusion. MDDCT administration was associated with increased expression of IL-2 in CD4+ and CD8+ T-cells at 15 and/or 30 days after the first MDDCT administration. Moreover, intracellular TNF and IFN-γ expression was significantly increased in CD4+ T-cells. The number of candidates that increased in vitro the cytokine levels in CD4+ and CD8+ T cells upon stimulation with Gag peptides from baseline to day 15 and from baseline to day 30 and day 120 after MDDCT was significant as compared to Gag unstimulated response. This was accompanied by an increasing trend in the frequency of polyfunctional T-cells over time, which was visible when considering both cells expressing two and three out of the three cytokines examined. Conclusions MDDC had a mature profile, and this MDDCT promoted in-vitro T-cell immune responses in HIV-infected patients undergoing long-term suppressive antiretroviral treatment. Trial registration NCT02961829: (Multi Interventional Study Exploring HIV-1 Residual Replication: a Step Towards HIV-1 Eradication and Sterilizing Cure, https://www.clinicaltrials.gov/ct2/show/NCT02961829, posted November 11th, 2016) Supplementary Information The online version contains supplementary material available at 10.1186/s12981-021-00426-z.
Collapse
|
16
|
Alameh MG, Weissman D, Pardi N. Messenger RNA-Based Vaccines Against Infectious Diseases. Curr Top Microbiol Immunol 2022; 440:111-145. [PMID: 32300916 DOI: 10.1007/82_2020_202] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In vitro-transcribed, messenger RNA-based infectious disease vaccines have the potential to successfully address many of the weaknesses of traditional vaccine platforms, such as the lack of potency and/or durability of vaccine protection, time-consuming, and expensive manufacturing, and, in some cases, safety issues. This optimism is fueled by a great deal of impressive recent data demonstrating that mRNA vaccines have many of the attributes that are necessary for a viable new vaccine class for human use. This review briefly describes mRNA vaccine types, discusses the most relevant and recent publications on infectious disease mRNA vaccines, and highlights the hurdles that need to be overcome to bring this promising novel vaccine modality to the clinic.
Collapse
Affiliation(s)
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Norbert Pardi
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
17
|
Yu AM, Tu MJ. Deliver the promise: RNAs as a new class of molecular entities for therapy and vaccination. Pharmacol Ther 2021; 230:107967. [PMID: 34403681 PMCID: PMC9477512 DOI: 10.1016/j.pharmthera.2021.107967] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/06/2021] [Accepted: 07/13/2021] [Indexed: 12/19/2022]
Abstract
The concepts of developing RNAs as new molecular entities for therapies have arisen again and again since the discoveries of antisense RNAs, direct RNA-protein interactions, functional noncoding RNAs, and RNA-directed gene editing. The feasibility was demonstrated with the development and utilization of synthetic RNA agents to selectively control target gene expression, modulate protein functions or alter the genome to manage diseases. Rather, RNAs are labile to degradation and cannot cross cell membrane barriers, making it hard to develop RNA medications. With the development of viable RNA technologies, such as chemistry and pharmaceutics, eight antisense oligonucleotides (ASOs) (fomivirsen, mipomersen, eteplirsen, nusinersen, inotersen, golodirsen, viltolarsen and casimersen), one aptamer (pegaptanib), and three small interfering RNAs (siRNAs) (patisiran, givosiran and lumasiran) have been approved by the United States Food and Drug Administration (FDA) for therapies, and two mRNA vaccines (BNT162b2 and mRNA-1273) under Emergency Use Authorization for the prevention of COVID-19. Therefore, RNAs have become a great addition to small molecules, proteins/antibodies, and cell-based modalities to improve the public health. In this article, we first summarize the general characteristics of therapeutic RNA agents, including chemistry, common delivery strategies, mechanisms of actions, and safety. By overviewing individual RNA medications and vaccines approved by the FDA and some agents under development, we illustrate the unique compositions and pharmacological actions of RNA products. A new era of RNA research and development will likely lead to commercialization of more RNA agents for medical use, expanding the range of therapeutic targets and increasing the diversity of molecular modalities.
Collapse
Affiliation(s)
- Ai-Ming Yu
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA.
| | - Mei-Juan Tu
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA
| |
Collapse
|
18
|
Rapaka RR, Cross AS, McArthur MA. Using Adjuvants to Drive T Cell Responses for Next-Generation Infectious Disease Vaccines. Vaccines (Basel) 2021; 9:vaccines9080820. [PMID: 34451945 PMCID: PMC8402546 DOI: 10.3390/vaccines9080820] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 12/12/2022] Open
Abstract
Using adjuvants to drive features of T cell responses to vaccine antigens is an important technological challenge in the design of new and improved vaccines against infections. Properties such as T helper cell function, T cell memory, and CD8+ T cell cytotoxicity may play critical roles in optimal and long-lived immunity through vaccination. Directly manipulating specific immune activation or antigen delivery pathways with adjuvants may selectively augment desired T cell responses in vaccination and may improve the effectiveness and durability of vaccine responses in humans. In this review we outline recently studied adjuvants in their potential for antigen presenting cell and T cell programming during vaccination, with an emphasis on what has been observed in studies in humans as available.
Collapse
|
19
|
To KKW, Cho WCS. An overview of rational design of mRNA-based therapeutics and vaccines. Expert Opin Drug Discov 2021; 16:1307-1317. [PMID: 34058918 DOI: 10.1080/17460441.2021.1935859] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Messenger RNA (mRNA)-based therapeutics and vaccines have emerged as a disruptive new drug class for various applications, including regenerative medicine, cancer treatment, and prophylactic and therapeutic vaccinations. AREAS COVERED This review provides an update about the rational structure-based design of various formats of mRNA-based therapeutics. The authors discuss the recent advances in the mRNA modifications that have been used to enhance stability, promote translation efficiency and regulate immunogenicity for specific applications. EXPERT OPINION Extensive research efforts have been made to optimize mRNA constructs and preparation procedures to unleash the full potential of mRNA-based therapeutics and vaccines. Sequence optimization (untranslated region and codon usage), chemical engineering of nucleotides and modified 5'cap, and optimization of in vitro transcription and mRNA purification protocols have overcome the major obstacles (instability, delivery, immunogenicity and safety) hindering the clinical applications of mRNA therapeutics and vaccines. The optimized design parameters should not be applied as default to different biological systems, but rather individually optimized for each mRNA sequence and intended application. Further advancement in the mRNA design and delivery technologies for achieving cell type- and organ site-specificity will broaden the scope and usefulness of this new class of drugs.
Collapse
Affiliation(s)
- Kenneth K W To
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - William C S Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, China
| |
Collapse
|
20
|
Esteban I, Pastor-Quiñones C, Usero L, Plana M, García F, Leal L. In the Era of mRNA Vaccines, Is There Any Hope for HIV Functional Cure? Viruses 2021; 13:501. [PMID: 33803790 PMCID: PMC8003302 DOI: 10.3390/v13030501] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/10/2021] [Accepted: 03/12/2021] [Indexed: 01/07/2023] Open
Abstract
Over 36 million people worldwide are infected with HIV. Antiretroviral therapy (ART) has proven to be highly effective to prevent HIV-1 transmission, clinical progression and death. Despite this success, the number of HIV-1 infected individuals continues increasing and ART should be taken for life. Therefore, there are two main priorities: the development of preventive vaccines to protect from HIV acquisition and achieve an efficient control of HIV infection in the absence of ART (functional cure). In this sense, in the last few years, there has been a broad interest in new and innovative approaches such as mRNA-based vaccines. RNA-based immunogens represent a promising alternative to conventional vaccines because of their high potency, capacity for rapid development and potential for low-cost manufacture and safe administration. Some mRNA-based vaccines platforms against infectious diseases have demonstrated encouraging results in animal models and humans. However, their application is still limited because the instability and inefficient in vivo delivery of mRNA. Immunogens, design, immunogenicity, chemical modifications on the molecule or the vaccine delivery methods are all crucial interventions for improvement. In this review we, will present the current knowledge and challenges in this research field. mRNA vaccines hold great promises as part of a combined strategy, for achieving HIV functional cure.
Collapse
Affiliation(s)
- Ignasi Esteban
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (I.E.); (C.P.-Q.); (L.U.); (M.P.); (F.G.)
| | - Carmen Pastor-Quiñones
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (I.E.); (C.P.-Q.); (L.U.); (M.P.); (F.G.)
| | - Lorena Usero
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (I.E.); (C.P.-Q.); (L.U.); (M.P.); (F.G.)
| | - Montserrat Plana
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (I.E.); (C.P.-Q.); (L.U.); (M.P.); (F.G.)
| | - Felipe García
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (I.E.); (C.P.-Q.); (L.U.); (M.P.); (F.G.)
- Infectious Diseases Department, Hospital Clínic, University of Barcelona, 08036 Barcelona, Spain
| | - Lorna Leal
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (I.E.); (C.P.-Q.); (L.U.); (M.P.); (F.G.)
- Infectious Diseases Department, Hospital Clínic, University of Barcelona, 08036 Barcelona, Spain
| |
Collapse
|
21
|
Heine A, Juranek S, Brossart P. Clinical and immunological effects of mRNA vaccines in malignant diseases. Mol Cancer 2021; 20:52. [PMID: 33722265 PMCID: PMC7957288 DOI: 10.1186/s12943-021-01339-1] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 02/23/2021] [Indexed: 12/12/2022] Open
Abstract
In vitro-transcribed messenger RNA-based therapeutics represent a relatively novel and highly efficient class of drugs. Several recently published studies emphasize the potential efficacy of mRNA vaccines in treating different types of malignant and infectious diseases where conventional vaccine strategies and platforms fail to elicit protective immune responses. mRNA vaccines have lately raised high interest as potent vaccines against SARS-CoV2. Direct application of mRNA or its electroporation into dendritic cells was shown to induce polyclonal CD4+ and CD8+ mediated antigen-specific T cell responses as well as the production of protective antibodies with the ability to eliminate transformed or infected cells. More importantly, the vaccine composition may include two or more mRNAs coding for different proteins or long peptides. This enables the induction of polyclonal immune responses against a broad variety of epitopes within the encoded antigens that are presented on various MHC complexes, thus avoiding the restriction to a certain HLA molecule or possible immune escape due to antigen-loss. The development and design of mRNA therapies was recently boosted by several critical innovations including the development of technologies for the production and delivery of high quality and stable mRNA. Several technical obstacles such as stability, delivery and immunogenicity were addressed in the past and gradually solved in the recent years.This review will summarize the most recent technological developments and application of mRNA vaccines in clinical trials and discusses the results, challenges and future directions with a special focus on the induced innate and adaptive immune responses.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Drug Delivery Systems
- Gene Expression Regulation, Neoplastic
- Gene Transfer Techniques
- Humans
- Immunity
- Immunotherapy
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/pathology
- Neoplasms/etiology
- Neoplasms/pathology
- Neoplasms/therapy
- RNA Stability
- RNA, Messenger/genetics
- RNA, Messenger/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Annkristin Heine
- Medical Clinic III for Oncology, Hematology, Immune-Oncology and Rheumatology, University Hospital Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Stefan Juranek
- Medical Clinic III for Oncology, Hematology, Immune-Oncology and Rheumatology, University Hospital Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Peter Brossart
- Medical Clinic III for Oncology, Hematology, Immune-Oncology and Rheumatology, University Hospital Bonn, Venusberg Campus 1, 53127, Bonn, Germany.
| |
Collapse
|
22
|
Cagigi A, Loré K. Immune Responses Induced by mRNA Vaccination in Mice, Monkeys and Humans. Vaccines (Basel) 2021; 9:61. [PMID: 33477534 PMCID: PMC7831080 DOI: 10.3390/vaccines9010061] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
In this concise review, we summarize the concepts behind mRNA vaccination. We discuss the innate and adaptive immune response generated by mRNA vaccines in different animal models and in humans. We give examples of viral infections where mRNA vaccines have shown to induce potent responses and we discuss in more detail the recent SARS-CoV-2 mRNA vaccine trials in humans.
Collapse
Affiliation(s)
| | - Karin Loré
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 161 64 Solna, Sweden;
| |
Collapse
|
23
|
Abstract
The first proof-of-concept studies about the feasibility of genetic vaccines were published over three decades ago, opening the way for future development. The idea of nonviral antigen delivery had multiple advantages over the traditional live or inactivated pathogen-based vaccines, but a great deal of effort had to be invested to turn the idea of genetic vaccination into reality. Although early proof-of-concept studies were groundbreaking, they also showed that numerous aspects of genetic vaccines needed to be improved. Until the early 2000s, the vast majority of effort was invested into the development of DNA vaccines due to the potential issues of instability and low in vivo translatability of messenger RNA (mRNA). In recent years, numerous studies have demonstrated the outstanding abilities of mRNA to elicit potent immune responses against infectious pathogens and different types of cancer, making it a viable platform for vaccine development. Multiple mRNA vaccine platforms have been developed and evaluated in small and large animals and humans and the results seem to be promising. RNA-based vaccines have important advantages over other vaccine approaches including outstanding efficacy, safety, and the potential for rapid, inexpensive, and scalable production. There is a substantial investment by new mRNA companies into the development of mRNA therapeutics, particularly vaccines, increasing the number of basic and translational research publications and human clinical trials underway. This review gives a broad overview about genetic vaccines and mainly focuses on the past and present of mRNA vaccines along with the future directions to bring this potent vaccine platform closer to therapeutic use.
Collapse
|
24
|
D'haese S, Lacroix C, Garcia F, Plana M, Ruta S, Vanham G, Verrier B, Aerts JL. Off the beaten path: Novel mRNA-nanoformulations for therapeutic vaccination against HIV. J Control Release 2020; 330:1016-1033. [PMID: 33181204 DOI: 10.1016/j.jconrel.2020.11.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 11/05/2020] [Accepted: 11/06/2020] [Indexed: 12/16/2022]
Abstract
Over the last few years, immunotherapy for HIV in general and therapeutic vaccination in particular, has received a tremendous boost, both in preclinical research and in clinical applications. This interest is based on the evidence that the immune system plays a crucial role in controlling HIV infection, as shown for long-term non-progressors and elite controllers, and that immune responses can be manipulated towards targeting conserved epitopes. So far, the most successful approach has been vaccination with autologous dendritic cells (DCs) loaded ex vivo with antigens and activation signals. Although this approach offers much promise, it also comes with significant drawbacks such as the requirement of a specialized infrastructure and expertise, as well as major challenges for logistics and storage, making it extremely time consuming and costly. Therefore, methods are being developed to avoid the use of ex vivo generated, autologous DCs. One of these methods is based on mRNA for therapeutic vaccination. mRNA has proven to be a very promising vaccine platform, as the coding information for any desired protein, including antigens and activation signals, can be generated in a very short period of time, showing promise both as an off-the-shelf therapy and as a personalized approach. However, an important drawback of this approach is the short half-life of native mRNA, due to the presence of ambient RNases. In addition, proper immunization requires that the antigens are expressed, processed and presented at the right immunological site (e.g. the lymphoid tissues). An ambivalent aspect of mRNA as a vaccine is its capacity to induce type I interferons, which can have beneficial adjuvant effects, but also deleterious effects on mRNA stability and translation. Thus, proper formulation of the mRNA is crucially important. Many approaches for RNA formulation have already been tested, with mixed success. In this review we discuss the state-of-the-art and future trends for mRNA-nanoparticle formulations for HIV vaccination, both in the prophylactic and in the therapeutic setting.
Collapse
Affiliation(s)
- Sigrid D'haese
- Neuro-Aging & Viro-Immunotherapy (NAVI), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Céline Lacroix
- Institute for the Biology and Chemistry of Proteins (IBCP), Lyon, France
| | | | | | - Simona Ruta
- Carol Davila University of Medicine and Pharmacy, Stefan S. Nicolau Institute of Virology, Bucharest, Romania
| | - Guido Vanham
- Institute of Tropical Medicine and University of Antwerp, Antwerp, Belgium
| | - Bernard Verrier
- Institute for the Biology and Chemistry of Proteins (IBCP), Lyon, France
| | - Joeri L Aerts
- Neuro-Aging & Viro-Immunotherapy (NAVI), Vrije Universiteit Brussel (VUB), Brussels, Belgium.
| |
Collapse
|
25
|
Bloom K, van den Berg F, Arbuthnot P. Self-amplifying RNA vaccines for infectious diseases. Gene Ther 2020; 28:117-129. [PMID: 33093657 PMCID: PMC7580817 DOI: 10.1038/s41434-020-00204-y] [Citation(s) in RCA: 215] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/29/2020] [Accepted: 10/08/2020] [Indexed: 12/28/2022]
Abstract
Vaccinology is shifting toward synthetic RNA platforms which allow for rapid, scalable, and cell-free manufacturing of prophylactic and therapeutic vaccines. The simple development pipeline is based on in vitro transcription of antigen-encoding sequences or immunotherapies as synthetic RNA transcripts, which are then formulated for delivery. This approach may enable a quicker response to emerging disease outbreaks, as is evident from the swift pursuit of RNA vaccine candidates for the global SARS-CoV-2 pandemic. Both conventional and self-amplifying RNAs have shown protective immunization in preclinical studies against multiple infectious diseases including influenza, RSV, Rabies, Ebola, and HIV-1. Self-amplifying RNAs have shown enhanced antigen expression at lower doses compared to conventional mRNA, suggesting this technology may improve immunization. This review will explore how self-amplifying RNAs are emerging as important vaccine candidates for infectious diseases, the advantages of synthetic manufacturing approaches, and their potential for preventing and treating chronic infections.
Collapse
Affiliation(s)
- Kristie Bloom
- Wits/SAMRC Antiviral Gene Therapy Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Private Bag 3, WITS, Johannesburg, 2050, South Africa.
| | - Fiona van den Berg
- Wits/SAMRC Antiviral Gene Therapy Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Private Bag 3, WITS, Johannesburg, 2050, South Africa
| | - Patrick Arbuthnot
- Wits/SAMRC Antiviral Gene Therapy Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Private Bag 3, WITS, Johannesburg, 2050, South Africa
| |
Collapse
|
26
|
Yu AM, Choi YH, Tu MJ. RNA Drugs and RNA Targets for Small Molecules: Principles, Progress, and Challenges. Pharmacol Rev 2020; 72:862-898. [PMID: 32929000 PMCID: PMC7495341 DOI: 10.1124/pr.120.019554] [Citation(s) in RCA: 202] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
RNA-based therapies, including RNA molecules as drugs and RNA-targeted small molecules, offer unique opportunities to expand the range of therapeutic targets. Various forms of RNAs may be used to selectively act on proteins, transcripts, and genes that cannot be targeted by conventional small molecules or proteins. Although development of RNA drugs faces unparalleled challenges, many strategies have been developed to improve RNA metabolic stability and intracellular delivery. A number of RNA drugs have been approved for medical use, including aptamers (e.g., pegaptanib) that mechanistically act on protein target and small interfering RNAs (e.g., patisiran and givosiran) and antisense oligonucleotides (e.g., inotersen and golodirsen) that directly interfere with RNA targets. Furthermore, guide RNAs are essential components of novel gene editing modalities, and mRNA therapeutics are under development for protein replacement therapy or vaccination, including those against unprecedented severe acute respiratory syndrome coronavirus pandemic. Moreover, functional RNAs or RNA motifs are highly structured to form binding pockets or clefts that are accessible by small molecules. Many natural, semisynthetic, or synthetic antibiotics (e.g., aminoglycosides, tetracyclines, macrolides, oxazolidinones, and phenicols) can directly bind to ribosomal RNAs to achieve the inhibition of bacterial infections. Therefore, there is growing interest in developing RNA-targeted small-molecule drugs amenable to oral administration, and some (e.g., risdiplam and branaplam) have entered clinical trials. Here, we review the pharmacology of novel RNA drugs and RNA-targeted small-molecule medications, with a focus on recent progresses and strategies. Challenges in the development of novel druggable RNA entities and identification of viable RNA targets and selective small-molecule binders are discussed. SIGNIFICANCE STATEMENT: With the understanding of RNA functions and critical roles in diseases, as well as the development of RNA-related technologies, there is growing interest in developing novel RNA-based therapeutics. This comprehensive review presents pharmacology of both RNA drugs and RNA-targeted small-molecule medications, focusing on novel mechanisms of action, the most recent progress, and existing challenges.
Collapse
MESH Headings
- Aptamers, Nucleotide/pharmacology
- Aptamers, Nucleotide/therapeutic use
- Betacoronavirus
- COVID-19
- Chemistry Techniques, Analytical/methods
- Chemistry Techniques, Analytical/standards
- Clustered Regularly Interspaced Short Palindromic Repeats
- Coronavirus Infections/drug therapy
- Drug Delivery Systems/methods
- Drug Development/organization & administration
- Drug Discovery
- Humans
- MicroRNAs/pharmacology
- MicroRNAs/therapeutic use
- Oligonucleotides, Antisense/pharmacology
- Oligonucleotides, Antisense/therapeutic use
- Pandemics
- Pneumonia, Viral/drug therapy
- RNA/adverse effects
- RNA/drug effects
- RNA/pharmacology
- RNA, Antisense/pharmacology
- RNA, Antisense/therapeutic use
- RNA, Messenger/drug effects
- RNA, Messenger/pharmacology
- RNA, Ribosomal/drug effects
- RNA, Ribosomal/pharmacology
- RNA, Small Interfering/pharmacology
- RNA, Small Interfering/therapeutic use
- RNA, Viral/drug effects
- Ribonucleases/metabolism
- Riboswitch/drug effects
- SARS-CoV-2
Collapse
Affiliation(s)
- Ai-Ming Yu
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, California (A.-M.Y., Y.H.C., M.-J.T.) and College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang-si, Gyonggi-do, Republic of Korea (Y.H.C.)
| | - Young Hee Choi
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, California (A.-M.Y., Y.H.C., M.-J.T.) and College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang-si, Gyonggi-do, Republic of Korea (Y.H.C.)
| | - Mei-Juan Tu
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, California (A.-M.Y., Y.H.C., M.-J.T.) and College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang-si, Gyonggi-do, Republic of Korea (Y.H.C.)
| |
Collapse
|
27
|
Affiliation(s)
- Chaoyang Meng
- Department of Nanomedicine Houston Methodist Research Institute Houston TX 77030 USA
- Xiangya Hospital of Central South University Changsha Hunan 410000 China
| | - Zhe Chen
- Department of Nanomedicine Houston Methodist Research Institute Houston TX 77030 USA
- Xiangya Hospital of Central South University Changsha Hunan 410000 China
| | - Gang Li
- Department of Nanomedicine Houston Methodist Research Institute Houston TX 77030 USA
| | - Thomas Welte
- Department of Nanomedicine Houston Methodist Research Institute Houston TX 77030 USA
| | - Haifa Shen
- Department of Nanomedicine Houston Methodist Research Institute Houston TX 77030 USA
- Cancer Center Houston Methodist Hospital Houston TX 77030 USA
- Department of Cell and Developmental Biology Weill Cornell Medical College New York NY 10065 USA
| |
Collapse
|
28
|
The Evolution of Dendritic Cell Immunotherapy against HIV-1 Infection: Improvements and Outlook. J Immunol Res 2020; 2020:9470102. [PMID: 32537473 PMCID: PMC7267878 DOI: 10.1155/2020/9470102] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 04/28/2020] [Indexed: 12/18/2022] Open
Abstract
Dendritic cells (DC) are key phagocytic cells that play crucial roles in both the innate and adaptive immune responses against the human immunodeficiency virus type 1 (HIV-1). By processing and presenting pathogen-derived antigens, dendritic cells initiate a directed response against infected cells. They activate the adaptive immune system upon recognition of pathogen-associated molecular patterns (PAMPs) on infected cells. During the course of HIV-1 infection, a successful adaptive (cytotoxic CD8+ T-cell) response is necessary for preventing the progression and spread of infection in a variety of cells. Dendritic cells have thus been recognized as a valuable tool in the development of immunotherapeutic approaches and vaccines effective against HIV-1. The advancements in dendritic cell vaccines in cancers have paved the way for applications of this form of immunotherapy to HIV-1 infection. Clinical trials with patients infected with HIV-1 who are well-suppressed by antiretroviral therapy (ART) were recently performed to assess the efficacy of DC vaccines, with the goal of mounting an HIV-1 antigen-specific T-cell response, ideally to clear infection and eliminate the need for long-term ART. This review summarizes and compares methods and efficacies of a number of DC vaccine trials utilizing autologous dendritic cells loaded with HIV-1 antigens. The potential for advancement and novel strategies of improving efficacy of this type of immunotherapy is also discussed.
Collapse
|
29
|
Ribonucleic Acid Engineering of Dendritic Cells for Therapeutic Vaccination: Ready 'N Able to Improve Clinical Outcome? Cancers (Basel) 2020; 12:cancers12020299. [PMID: 32012714 PMCID: PMC7072269 DOI: 10.3390/cancers12020299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/06/2020] [Accepted: 01/19/2020] [Indexed: 02/06/2023] Open
Abstract
Targeting and exploiting the immune system has become a valid alternative to conventional options for treating cancer and infectious disease. Dendritic cells (DCs) take a central place given their role as key orchestrators of immunity. Therapeutic vaccination with autologous DCs aims to stimulate the patient's own immune system to specifically target his/her disease and has proven to be an effective form of immunotherapy with very little toxicity. A great amount of research in this field has concentrated on engineering these DCs through ribonucleic acid (RNA) to improve vaccine efficacy and thereby the historically low response rates. We reviewed in depth the 52 clinical trials that have been published on RNA-engineered DC vaccination, spanning from 2001 to date and reporting on 696 different vaccinated patients. While ambiguity prevents reliable quantification of effects, these trials do provide evidence that RNA-modified DC vaccination can induce objective clinical responses and survival benefit in cancer patients through stimulation of anti-cancer immunity, without significant toxicity. Succinct background knowledge of RNA engineering strategies and concise conclusions from available clinical and recent preclinical evidence will help guide future research in the larger domain of DC immunotherapy.
Collapse
|
30
|
Therapeutic Cancer Vaccination with Ex Vivo RNA-Transfected Dendritic Cells-An Update. Pharmaceutics 2020; 12:pharmaceutics12020092. [PMID: 31979205 PMCID: PMC7076681 DOI: 10.3390/pharmaceutics12020092] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/09/2020] [Accepted: 01/18/2020] [Indexed: 12/19/2022] Open
Abstract
Over the last two decades, dendritic cell (DC) vaccination has been studied extensively as active immunotherapy in cancer treatment and has been proven safe in all clinical trials both with respect to short and long-term side effects. For antigen-loading of dendritic cells (DCs) one method is to introduce mRNA coding for the desired antigens. To target the whole antigenic repertoire of a tumor, even the total tumor mRNA of a macrodissected biopsy sample can be used. To date, reports have been published on a total of 781 patients suffering from different tumor entities and HIV-infection, who have been treated with DCs loaded with mRNA. The majority of those were melanoma patients, followed by HIV-infected patients, but leukemias, brain tumors, prostate cancer, renal cell carcinomas, pancreatic cancers and several others have also been treated. Next to antigen-loading, mRNA-electroporation allows a purposeful manipulation of the DCs’ phenotype and function to enhance their immunogenicity. In this review, we intend to give a comprehensive summary of what has been published regarding clinical testing of ex vivo generated mRNA-transfected DCs, with respect to safety and risk/benefit evaluations, choice of tumor antigens and RNA-source, and the design of better DCs for vaccination by transfection of mRNA-encoded functional proteins.
Collapse
|
31
|
da Silva LT, da Silva WC, de Almeida A, da Silva Reis D, Santillo BT, Rigato PO, da Silva Duarte AJ, Oshiro TM. Characterization of monocyte-derived dendritic cells used in immunotherapy for HIV-1-infected individuals. Immunotherapy 2019; 10:871-885. [PMID: 30073900 DOI: 10.2217/imt-2017-0165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
AIMS A therapeutic vaccine based on monocyte-derived dendritic cells (MDDCs) has been shown to represent a promising strategy for the treatment of cancer and viral infections. Here, we characterized the MDDCs used as an immunogen in a clinical trial for an anti-HIV-1 therapeutic vaccine. PATIENTS & METHODS Monocytes obtained from 17 HIV-infected individuals were differentiated into MDDCs and, after loading with autologous HIV, the cells were characterized concerning surface molecule expression, migratory and phagocytosis capacity, cytokine production and the induction of an effective cell-mediated immune response. RESULTS The MDDCs were able to induce antigen-specific responses in autologous CD4+ and CD8+ T lymphocytes. CONCLUSIONS Despite a large interindividual variability, the results suggested that MDDCs present the potential to promote immune responses in vaccinated patients.
Collapse
Affiliation(s)
- Laís Teodoro da Silva
- Laboratorio de Investigacao em Dermatologia e Imunodeficiencias, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR. 05403-903, Brazil
| | - Wanessa Cardoso da Silva
- Laboratorio de Investigacao em Dermatologia e Imunodeficiencias, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR. 05403-903, Brazil
| | - Alexandre de Almeida
- Laboratorio de Investigacao em Dermatologia e Imunodeficiencias, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR. 05403-903, Brazil
| | - Denise da Silva Reis
- Laboratorio de Investigacao em Dermatologia e Imunodeficiencias, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR. 05403-903, Brazil
| | - Bruna Tereso Santillo
- Laboratorio de Investigacao em Dermatologia e Imunodeficiencias, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR. 05403-903, Brazil
| | | | - Alberto José da Silva Duarte
- Laboratorio de Investigacao em Dermatologia e Imunodeficiencias, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR. 05403-903, Brazil
| | - Telma Miyuki Oshiro
- Laboratorio de Investigacao em Dermatologia e Imunodeficiencias, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR. 05403-903, Brazil
| |
Collapse
|
32
|
de Jong W, Aerts J, Allard S, Brander C, Buyze J, Florence E, van Gorp E, Vanham G, Leal L, Mothe B, Thielemans K, Plana M, Garcia F, Gruters R. iHIVARNA phase IIa, a randomized, placebo-controlled, double-blinded trial to evaluate the safety and immunogenicity of iHIVARNA-01 in chronically HIV-infected patients under stable combined antiretroviral therapy. Trials 2019; 20:361. [PMID: 31208472 PMCID: PMC6580477 DOI: 10.1186/s13063-019-3409-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 05/06/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND HIV therapeutic vaccination aims to improve the immune responses against HIV in order to control viral replication without the need for combined antiretroviral therapy (cART). iHIVARNA-01 is a novel vaccine combining mRNA delivery and T-cell immunogen (HTI) based on conserved targets of effective antiviral T-cell responses. In addition, it holds adequate stimuli required for activating antigen presenting cells (APC)s and co-activating specific T-cells (TriMix), including human CD40L, constitutively active TLR4 (caTLR4) and CD70. We propose that in-vivo targeting of dendritic cells (DCs) by direct administration of a HIV mRNA encoding these immune modulating proteins might be an attractive alternative to target DCs in vitro. METHODS/DESIGN This is a phase-IIa, randomized, double-blinded, placebo-controlled, multicenter study in chronically HIV-1 infected patients under stable cART. One of the three study arms is randomly allocated to subjects. Three vaccinations with either HIVACAT T-cell immunogen (HTI)-TriMix (iHIVARNA-01), TriMix or water for injection (WFI) (weeks 0, 2 and 4) are administered by intranodal injection in the inguinal region. Two weeks after the last immunization (week 6) cART is stopped for 12 weeks. The two primary endpoints are: (1) safety and tolerability of intranodal iHIVARNA-01 vaccination compared with TriMix or WFI and (2) induced immunogenicity, i.e., increase in the frequency of HIV-specific T-cell responses between baseline, week 6 and 12 weeks after treatment interruption in iHIVARNA-01-treated patients as compared to the control groups, immunized with TriMix-mRNA or WFI measured by an IFNγ ELISPOT assay. Secondary endpoints include the evaluation of time to viral rebound, plasma viral load (pVL) at w18, the proportion of patients with control of viral load, induction of T-cell responses to new HIV epitopes, polyfunctionality of HIV-specific T-cells, CD8+ T-cell in-vitro HIV suppressive capacity, the effect on viral reservoir (measured by proviral DNA and cell-associated RNA), assessment of viral immune escape by mutation and mRNA expression profiles of host immune genes. DISCUSSION This trial aims to direct target DC in situ with mRNA encoding HTI and TriMix for co-stimulation. Intranodal injection circumvents laborious DC isolation and handling in the laboratory. The trial extends on the safety results of a phase-I dose-escalating trial. This candidate vaccine could complement or even replace cART for chronic HIV infection and could be applicable to improve the care and cost of HIV infection. TRIAL REGISTRATION EudraCT 2016-002724-83 (22 September 2016); ClinicalTrials.gov, ID: NCT02888756 . Registered on 23 August 2016.
Collapse
Affiliation(s)
- Wesley de Jong
- Department of Viroscience, Erasmus MC, Room Ee-1726, P.O. Box 2040, 3000, CA, Rotterdam, The Netherlands
| | - Joeri Aerts
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sabine Allard
- Department of Internal Medicine and Infectious Diseases, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Christian Brander
- Infectious Diseases Unit, IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Badalona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.,AELIX Therapeutics, Parc Científic de Barcelona, Barcelona, Spain.,University of Vic - Central University of Catalonia (UVic-UCC), Vic, Spain
| | - Jozefien Buyze
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine and, Antwerp, Belgium.,Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Eric Florence
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine and, Antwerp, Belgium.,Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Eric van Gorp
- Department of Viroscience, Erasmus MC, Room Ee-1726, P.O. Box 2040, 3000, CA, Rotterdam, The Netherlands
| | - Guido Vanham
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine and, Antwerp, Belgium.,Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Lorna Leal
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Villarroel, 170, 08036, Barcelona, Spain.,Infectious Diseases Unit, Hospital Clínic, Villarroel, 170, 08036, Barcelona, Spain
| | - Beatriz Mothe
- Infectious Diseases Unit, IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Badalona, Spain.,University of Vic - Central University of Catalonia (UVic-UCC), Vic, Spain
| | - Kris Thielemans
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Montse Plana
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Villarroel, 170, 08036, Barcelona, Spain
| | - Félipe Garcia
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Villarroel, 170, 08036, Barcelona, Spain. .,Infectious Diseases Unit, Hospital Clínic, Villarroel, 170, 08036, Barcelona, Spain.
| | - Rob Gruters
- Department of Viroscience, Erasmus MC, Room Ee-1726, P.O. Box 2040, 3000, CA, Rotterdam, The Netherlands.
| | | |
Collapse
|
33
|
da Silva LT, Santillo BT, de Almeida A, Duarte AJDS, Oshiro TM. Using Dendritic Cell-Based Immunotherapy to Treat HIV: How Can This Strategy be Improved? Front Immunol 2018; 9:2993. [PMID: 30619346 PMCID: PMC6305438 DOI: 10.3389/fimmu.2018.02993] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 12/04/2018] [Indexed: 11/13/2022] Open
Abstract
Harnessing dendritic cells (DC) to treat HIV infection is considered a key strategy to improve anti-HIV treatment and promote the discovery of functional or sterilizing cures. Although this strategy represents a promising approach, the results of currently published trials suggest that opportunities to optimize its performance still exist. In addition to the genetic and clinical characteristics of patients, the efficacy of DC-based immunotherapy depends on the quality of the vaccine product, which is composed of precursor-derived DC and an antigen for pulsing. Here, we focus on some factors that can interfere with vaccine production and should thus be considered to improve DC-based immunotherapy for HIV infection.
Collapse
Affiliation(s)
- Laís Teodoro da Silva
- Laboratorio de Investigacao em Dermatologia e Imunodeficiencias, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Bruna Tereso Santillo
- Laboratorio de Investigacao em Dermatologia e Imunodeficiencias, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Alexandre de Almeida
- Laboratorio de Investigacao em Dermatologia e Imunodeficiencias, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Alberto Jose da Silva Duarte
- Laboratorio de Investigacao em Dermatologia e Imunodeficiencias, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Telma Miyuki Oshiro
- Laboratorio de Investigacao em Dermatologia e Imunodeficiencias, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
34
|
Xu Y, Huang L, Kirschman JL, Vanover DA, Tiwari PM, Santangelo PJ, Shen X, Russell DG. Exploitation of Synthetic mRNA To Drive Immune Effector Cell Recruitment and Functional Reprogramming In Vivo. THE JOURNAL OF IMMUNOLOGY 2018; 202:608-617. [PMID: 30541883 DOI: 10.4049/jimmunol.1800924] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 11/05/2018] [Indexed: 12/24/2022]
Abstract
Therapeutic strategies based on in vitro-transcribed mRNA (IVT) are attractive because they avoid the permanent signature of genomic integration that is associated with DNA-based therapy and result in the transient production of proteins of interest. To date, IVT has mainly been used in vaccination protocols to generate immune responses to foreign Ags. In this "proof-of-principle" study, we explore a strategy of combinatorial IVT to recruit and reprogram immune effector cells to acquire divergent biological functions in mice in vivo. First, we demonstrate that synthetic mRNA encoding CCL3 is able to recruit murine monocytes in a nonprogrammed state, exhibiting neither bactericidal nor tissue-repairing properties. However, upon addition of either Ifn-γ mRNA or Il-4 mRNA, we successfully polarized these cells to adopt either M1 or M2 macrophage activation phenotypes. This cellular reprogramming was demonstrated through increased expression of known surface markers and through the differential modulation of NADPH oxidase activity, or the superoxide burst. Our study demonstrates how IVT strategies can be combined to recruit and reprogram immune effector cells that have the capacity to fulfill complex biological tasks in vivo.
Collapse
Affiliation(s)
- Yitian Xu
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853.,Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853
| | - Lu Huang
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853
| | - Jonathan L Kirschman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332
| | - Daryll A Vanover
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332
| | - Pooja M Tiwari
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332
| | - Philip J Santangelo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332
| | - Xiling Shen
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853.,School of Electrical and Computer Engineering, Cornell University, Ithaca, NY 14853; and.,Department of Biomedical Engineering, Duke University, Durham, NC 27708
| | - David G Russell
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853;
| |
Collapse
|
35
|
Shin H, Park SJ, Yim Y, Kim J, Choi C, Won C, Min DH. Recent Advances in RNA Therapeutics and RNA Delivery Systems Based on Nanoparticles. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800065] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Hojeong Shin
- Center for RNA Research; Institute for Basic Science; Seoul National University; Seoul 08826 Republic of Korea
- Department of Chemistry; Seoul National University; Seoul 08826 Republic of Korea
| | - Se-Jin Park
- Center for RNA Research; Institute for Basic Science; Seoul National University; Seoul 08826 Republic of Korea
- Department of Chemistry; Seoul National University; Seoul 08826 Republic of Korea
| | - Yeajee Yim
- Center for RNA Research; Institute for Basic Science; Seoul National University; Seoul 08826 Republic of Korea
- Department of Chemistry; Seoul National University; Seoul 08826 Republic of Korea
| | - Jungho Kim
- Department of Chemistry; Seoul National University; Seoul 08826 Republic of Korea
- Institute of Biotherapeutics Convergence Technology; Lemonex Inc.; Seoul 08826 Republic of Korea
| | - Chulwon Choi
- Center for RNA Research; Institute for Basic Science; Seoul National University; Seoul 08826 Republic of Korea
- Department of Chemistry; Seoul National University; Seoul 08826 Republic of Korea
| | - Cheolhee Won
- Institute of Biotherapeutics Convergence Technology; Lemonex Inc.; Seoul 08826 Republic of Korea
| | - Dal-Hee Min
- Center for RNA Research; Institute for Basic Science; Seoul National University; Seoul 08826 Republic of Korea
- Department of Chemistry; Seoul National University; Seoul 08826 Republic of Korea
- Institute of Biotherapeutics Convergence Technology; Lemonex Inc.; Seoul 08826 Republic of Korea
| |
Collapse
|
36
|
Meng Z, O'Keeffe-Ahern J, Lyu J, Pierucci L, Zhou D, Wang W. A new developing class of gene delivery: messenger RNA-based therapeutics. Biomater Sci 2018; 5:2381-2392. [PMID: 29063914 DOI: 10.1039/c7bm00712d] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Gene therapy has long been held as having the potential to become a front line treatment for various genetic disorders. However, the direct delivery of nucleic acids to correct a genetic disorder has numerous limitations owing to the inability of naked nucleic acids (DNA and RNA) to traverse the cell membrane. Recently, messenger RNA (mRNA) based delivery has become a more attractive alternative to DNA due to the relatively easier transfection process, higher efficiency and safety profile. As with all gene therapies, the central challenge that remains is the efficient delivery of nucleic acids intracellularly. This review presents the recent progress in mRNA delivery, focusing on comparing the advantages and limitations of non-viral based delivery vectors.
Collapse
Affiliation(s)
- Zhao Meng
- School of Materials Science and Engineering, Tianjin University, Tianjin 300072, China
| | | | | | | | | | | |
Collapse
|
37
|
Raja MAZ, Asma K, Aslam MS. Bio-inspired computational heuristics to study models of HIV infection of CD4+ T-cell. INT J BIOMATH 2018. [DOI: 10.1142/s1793524518500195] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
In this work, biologically-inspired computing framework is developed for HIV infection of CD4[Formula: see text] T-cell model using feed-forward artificial neural networks (ANNs), genetic algorithms (GAs), sequential quadratic programming (SQP) and hybrid approach based on GA-SQP. The mathematical model for HIV infection of CD4[Formula: see text] T-cells is represented with the help of initial value problems (IVPs) based on the system of ordinary differential equations (ODEs). The ANN model for the system is constructed by exploiting its strength of universal approximation. An objective function is developed for the system through unsupervised error using ANNs in the mean square sense. Training with weights of ANNs is carried out with GAs for effective global search supported with SQP for efficient local search. The proposed scheme is evaluated on a number of scenarios for the HIV infection model by taking the different levels for infected cells, natural substitution rates of uninfected cells, and virus particles. Comparisons of the approximate solutions are made with results of Adams numerical solver to establish the correctness of the proposed scheme. Accuracy and convergence of the approach are validated through the results of statistical analysis based on the sufficient large number of independent runs.
Collapse
Affiliation(s)
- Muhammad Asif Zahoor Raja
- Department of Electrical Engineering, COMSATS Institute of Information Technology, Attock Campus, Attock, Pakistan
| | - Kiran Asma
- Department of Computer Sciences, COMSATS Institute of Information Technology, Attock Campus, Attock, Pakistan
| | - Muhammad Saeed Aslam
- Pakistan Institute of Engineering and Applied Sciences, Nilore Islamabad, Pakistan
| |
Collapse
|
38
|
Pardi N, Hogan MJ, Porter FW, Weissman D. mRNA vaccines - a new era in vaccinology. Nat Rev Drug Discov 2018; 17:261-279. [PMID: 29326426 DOI: 10.1038/nrd.2017.243] [Citation(s) in RCA: 2438] [Impact Index Per Article: 406.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
mRNA vaccines represent a promising alternative to conventional vaccine approaches because of their high potency, capacity for rapid development and potential for low-cost manufacture and safe administration. However, their application has until recently been restricted by the instability and inefficient in vivo delivery of mRNA. Recent technological advances have now largely overcome these issues, and multiple mRNA vaccine platforms against infectious diseases and several types of cancer have demonstrated encouraging results in both animal models and humans. This Review provides a detailed overview of mRNA vaccines and considers future directions and challenges in advancing this promising vaccine platform to widespread therapeutic use.
Collapse
Affiliation(s)
- Norbert Pardi
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Michael J Hogan
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Frederick W Porter
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
39
|
Reap EA, Suryadevara CM, Batich KA, Sanchez-Perez L, Archer GE, Schmittling RJ, Norberg PK, Herndon JE, Healy P, Congdon KL, Gedeon PC, Campbell OC, Swartz AM, Riccione KA, Yi JS, Hossain-Ibrahim MK, Saraswathula A, Nair SK, Dunn-Pirio AM, Broome TM, Weinhold KJ, Desjardins A, Vlahovic G, McLendon RE, Friedman AH, Friedman HS, Bigner DD, Fecci PE, Mitchell DA, Sampson JH. Dendritic Cells Enhance Polyfunctionality of Adoptively Transferred T Cells That Target Cytomegalovirus in Glioblastoma. Cancer Res 2018; 78:256-264. [PMID: 29093005 PMCID: PMC5754236 DOI: 10.1158/0008-5472.can-17-0469] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 06/27/2017] [Accepted: 10/26/2017] [Indexed: 12/13/2022]
Abstract
Median survival for glioblastoma (GBM) remains <15 months. Human cytomegalovirus (CMV) antigens have been identified in GBM but not normal brain, providing an unparalleled opportunity to subvert CMV antigens as tumor-specific immunotherapy targets. A recent trial in recurrent GBM patients demonstrated the potential clinical benefit of adoptive T-cell therapy (ATCT) of CMV phosphoprotein 65 (pp65)-specific T cells. However, ex vivo analyses from this study found no change in the capacity of CMV pp65-specific T cells to gain multiple effector functions or polyfunctionality, which has been associated with superior antitumor efficacy. Previous studies have shown that dendritic cells (DC) could further enhance tumor-specific CD8+ T-cell polyfunctionality in vivo when administered as a vaccine. Therefore, we hypothesized that vaccination with CMV pp65 RNA-loaded DCs would enhance the frequency of polyfunctional CMV pp65-specific CD8+ T cells after ATCT. Here, we report prospective results of a pilot trial in which 22 patients with newly diagnosed GBM were initially enrolled, of which 17 patients were randomized to receive CMV pp65-specific T cells with CMV-DC vaccination (CMV-ATCT-DC) or saline (CMV-ATCT-saline). Patients who received CMV-ATCT-DC vaccination experienced a significant increase in the overall frequencies of IFNγ+, TNFα+, and CCL3+ polyfunctional, CMV-specific CD8+ T cells. These increases in polyfunctional CMV-specific CD8+ T cells correlated (R = 0.7371, P = 0.0369) with overall survival, although we cannot conclude this was causally related. Our data implicate polyfunctional T-cell responses as a potential biomarker for effective antitumor immunotherapy and support a formal assessment of this combination approach in a larger randomized study.Significance: A randomized pilot trial in patients with GBM implicates polyfunctional T-cell responses as a biomarker for effective antitumor immunotherapy. Cancer Res; 78(1); 256-64. ©2017 AACR.
Collapse
Affiliation(s)
- Elizabeth A Reap
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
| | - Carter M Suryadevara
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Kristen A Batich
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Luis Sanchez-Perez
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
| | - Gary E Archer
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Robert J Schmittling
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
| | - Pamela K Norberg
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
| | - James E Herndon
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, North Carolina
| | - Patrick Healy
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, North Carolina
| | - Kendra L Congdon
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
| | - Patrick C Gedeon
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
| | - Olivia C Campbell
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
| | - Adam M Swartz
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Katherine A Riccione
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
| | - John S Yi
- Division of Surgical Sciences, Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Mohammed K Hossain-Ibrahim
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
| | - Anirudh Saraswathula
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
| | - Smita K Nair
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
- Division of Surgical Sciences, Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Anastasie M Dunn-Pirio
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
| | - Taylor M Broome
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
| | - Kent J Weinhold
- Division of Surgical Sciences, Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Annick Desjardins
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
- Department of Neurology, Duke University Medical Center, Durham, North Carolina
| | - Gordana Vlahovic
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
| | - Roger E McLendon
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Allan H Friedman
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
| | - Henry S Friedman
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
| | - Darell D Bigner
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Peter E Fecci
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Duane A Mitchell
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - John H Sampson
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
40
|
Leal L, Lucero C, Gatell JM, Gallart T, Plana M, García F. New challenges in therapeutic vaccines against HIV infection. Expert Rev Vaccines 2017; 16:587-600. [PMID: 28431490 DOI: 10.1080/14760584.2017.1322513] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION There is a growing interest in developing curative strategies for HIV infection. Therapeutic vaccines are one of the most promising approaches. We will review the current knowledge and the new challenges in this research field. Areas covered: PubMed and ClinicalTrial.gov databases were searched to review the progress and prospects for clinical development of immunotherapies aimed to cure HIV infection. Dendritic cells (DC)-based vaccines have yielded the best results in the field. However, major immune-virologic barriers may hamper current vaccine strategies. We will focus on some new challenges as the antigen presentation by DCs, CTL escape mutations, B cell follicle sanctuary, host immune environment (inflammation, immune activation, tolerance), latent reservoir and the lack of surrogate markers of response. Finally, we will review the rationale for designing new therapeutic vaccine candidates to be used alone or in combination with other strategies to improve their effectiveness. Expert commentary: In the next future, the combination of DCs targeting candidates, inserts to redirect responses to unmutated parts of the virus, adjuvants to redirect responses to sanctuaries or improve the balance between activation/tolerance (IL-15, anti-PD1 antibodies) and latency reversing agents could be necessary to finally achieve the remission of HIV-1 infection.
Collapse
Affiliation(s)
- Lorna Leal
- a Infectious Diseases Unit, HIVACAT, Hospital Clínic, IDIBAPS , University of Barcelona , Barcelona , Spain
| | - Constanza Lucero
- a Infectious Diseases Unit, HIVACAT, Hospital Clínic, IDIBAPS , University of Barcelona , Barcelona , Spain
| | - Josep M Gatell
- a Infectious Diseases Unit, HIVACAT, Hospital Clínic, IDIBAPS , University of Barcelona , Barcelona , Spain
| | - Teresa Gallart
- b Retrovirology and Viral Immunopathology Laboratories, HIVACAT, Hospital Clínic, IDIBAPS , University of Barcelona , Barcelona , Spain
| | - Montserrat Plana
- b Retrovirology and Viral Immunopathology Laboratories, HIVACAT, Hospital Clínic, IDIBAPS , University of Barcelona , Barcelona , Spain
| | - Felipe García
- a Infectious Diseases Unit, HIVACAT, Hospital Clínic, IDIBAPS , University of Barcelona , Barcelona , Spain
| |
Collapse
|
41
|
Bartneck M. Immunomodulatory Nanomedicine. Macromol Biosci 2017; 17. [DOI: 10.1002/mabi.201700021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 02/21/2017] [Indexed: 12/28/2022]
Affiliation(s)
- Matthias Bartneck
- Department of Medicine III; Medical Faculty; RWTH Aachen; Pauwelsstr. 30 52074 Aachen Germany
| |
Collapse
|
42
|
Almeida CA, van Miert P, O'Driscoll K, Zoet YM, Chopra A, Watson M, de Santis D, Witt C, John M, Claas FHJ, D'Orsogna LJ. Stimulation of HIV-specific T cell clonotypes using allogeneic HLA. Cell Immunol 2017; 316:32-40. [PMID: 28372798 DOI: 10.1016/j.cellimm.2017.03.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 03/15/2017] [Accepted: 03/25/2017] [Indexed: 01/26/2023]
Abstract
We hypothesized that HIV-specific CD8 T cell clonotypes can be stimulated by allogeneic HLA molecules. Multiple HIV-specific CD8 T cell clones were derived from 12 individuals with chronic HIV infection, specific for 13 different HIV Gag antigens and restricted to 7 different HLA molecules. The generated T cell clones were assayed for alloreactivity against a panel of single HLA class I expressing cell lines (SALs). HIV-specific T cells recognising at least one allogeneic HLA molecule could be identified from 7 of 12 patients tested. Allorecognition was associated with IFNγ cytokine production, CD137 upregulation and cytotoxicity, suggesting high avidity allo-stimulation. Allo-HLA recognition by HIV-specific T cells was specific to the HIV target peptide/HLA restriction and TCR TRBV usage of the T cells. HIV-specific T cells do crossreact against allogeneic HLA molecules in an epitope and TRBV specific manner. Therefore allo-HLA stimulation could be exploited to induce or augment HIV-specific T cell responses.
Collapse
Affiliation(s)
- Coral-Ann Almeida
- Department of Clinical Immunology, Fiona Stanley Hospital, Perth, Australia; Pathwest Laboratory Medicine, Perth, Australia; Pathology and Laboratory Medicine, University of Western Australia, Perth, Australia
| | - Paula van Miert
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Centre, Leiden, The Netherlands
| | - Kane O'Driscoll
- Pathology and Laboratory Medicine, University of Western Australia, Perth, Australia
| | - Yvonne M Zoet
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Centre, Leiden, The Netherlands
| | - Abha Chopra
- Institute for Immunology and Infectious Diseases, Murdoch University, Perth, Australia
| | - Mark Watson
- Institute for Immunology and Infectious Diseases, Murdoch University, Perth, Australia
| | - Dianne de Santis
- Department of Clinical Immunology, Fiona Stanley Hospital, Perth, Australia; Pathwest Laboratory Medicine, Perth, Australia
| | - Campbell Witt
- Pathwest Laboratory Medicine, Perth, Australia; Department of Clinical Immunology, Royal Perth Hospital, Perth, Australia
| | - Mina John
- Pathwest Laboratory Medicine, Perth, Australia; Institute for Immunology and Infectious Diseases, Murdoch University, Perth, Australia; Department of Clinical Immunology, Royal Perth Hospital, Perth, Australia
| | - Frans H J Claas
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Centre, Leiden, The Netherlands
| | - Lloyd J D'Orsogna
- Department of Clinical Immunology, Fiona Stanley Hospital, Perth, Australia; Pathwest Laboratory Medicine, Perth, Australia; Pathology and Laboratory Medicine, University of Western Australia, Perth, Australia; Institute for Immunology and Infectious Diseases, Murdoch University, Perth, Australia.
| |
Collapse
|
43
|
Sergeeva OV, Koteliansky VE, Zatsepin TS. mRNA-Based Therapeutics - Advances and Perspectives. BIOCHEMISTRY (MOSCOW) 2017; 81:709-22. [PMID: 27449617 DOI: 10.1134/s0006297916070075] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
In this review we discuss features of mRNA synthesis and modifications used to minimize immune response and prolong efficiency of the translation process in vivo. Considerable attention is given to the use of liposomes and nanoparticles containing lipids and polymers for the mRNA delivery. Finally we briefly discuss mRNAs which are currently in the clinical trials for cancer immunotherapy, vaccination against infectious diseases, and replacement therapy.
Collapse
Affiliation(s)
- O V Sergeeva
- Lomonosov Moscow State University, Department of Chemistry, Moscow, 119991, Russia.
| | | | | |
Collapse
|
44
|
Gerer KF, Hoyer S, Dörrie J, Schaft N. Electroporation of mRNA as Universal Technology Platform to Transfect a Variety of Primary Cells with Antigens and Functional Proteins. Methods Mol Biol 2017; 1499:165-178. [PMID: 27987149 DOI: 10.1007/978-1-4939-6481-9_10] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Electroporation (EP) of mRNA into human cells is a broadly applicable method to transiently express proteins of choice in a variety of different cell types. We have spent more than a decade to optimize and adapt this method, first for antigen-loading of dendritic cells (DCs), and subsequently for T cells, B cells, bulk PBMCs, and several cell lines. In this regard, antigens were introduced, processed, and presented in context of MHC class I and II. Next to that, functional proteins like adhesion receptors, T-cell receptors (TCRs), chimeric antigen receptors (CARs), constitutively active signal transducers, and others were successfully expressed. We have also established this protocol under full GMP compliance as part of a manufacturing license to produce mRNA-electroporated DCs for therapeutic vaccination in clinical trials. Therefore, we here want to share our universal mRNA electroporation protocol and the experience we have gathered with this method. The advantages of the transfection method presented here are: (1) easy adaptation to different cell types, (2) scalability from 106 to approximately 108 cells per shot, (3) high transfection efficiency (80-99 %), (4) homogenous protein expression, (5) GMP compliance if the EP is performed in a class A clean room, and (6) no transgene integration into the genome. The provided protocol involves: Opti-MEM® as EP medium, a square-wave pulse with 500 V, and 4 mm cuvettes. To adapt the protocol to differently sized cells, simply the pulse time is altered. Next to the basic protocol, we also provide an extensive list of hints and tricks, which in our opinion are of great value for everyone who intends to use this transfection technique.
Collapse
Affiliation(s)
- Kerstin F Gerer
- Department of Dermatology, Universitätsklinikum Erlangen, Research campus, Hartmannstraße 14, 91052, Erlangen, Germany
| | - Stefanie Hoyer
- Department of Dermatology, Universitätsklinikum Erlangen, Research campus, Hartmannstraße 14, 91052, Erlangen, Germany
| | - Jan Dörrie
- Department of Dermatology, Universitätsklinikum Erlangen, Research campus, Hartmannstraße 14, 91052, Erlangen, Germany
| | - Niels Schaft
- Department of Dermatology, Universitätsklinikum Erlangen, Research campus, Hartmannstraße 14, 91052, Erlangen, Germany.
| |
Collapse
|
45
|
Abstract
In recent years, numerous studies have demonstrated the outstanding abilities of mRNA to elicit potent immune responses against pathogens, making it a viable new platform for vaccine development (reviewed in Weissman, Expert Rev Vaccines 14:265-281, 2015; Sahin et al., Nat Rev Drug Discov 13:759-780, 2014). The incorporation of modified nucleosides in mRNA has many advantages and is currently undergoing a renaissance in the field of therapeutic protein delivery. Its use in a vaccine against infectious diseases has only begun to be described, but offers advantages for the generation of potent and long-lived antibody responses. FPLC purification and substitution of modified nucleosides in the mRNA make it non-inflammatory and highly translatable (Kariko et al., Immunity 23:165-175, 2005; Kariko et al., Mol Ther 16:1833-1840, 2008; Kariko et al., Nucleic Acids Research 39:e142, 2011) that are crucial features for therapeutic relevance. Formulation of the mRNA in lipid nanoparticles (LNPs) protects it from degradation enabling high levels of protein production for extended periods of time (Pardi et al., J Control Release, 2015). Here, we describe a simple vaccination method using LNP-encapsulated 1-methylpseudouridine-containing FPLC purified mRNA in mice. Furthermore, we describe the evaluation of antigen-specific T and B cell responses elicited by this vaccine format.
Collapse
Affiliation(s)
- Norbert Pardi
- Department of Medicine, University of Pennsylvania, 52 Johnson Pavilion, Philadelphia, PA, 19104, USA
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania, 52 Johnson Pavilion, Philadelphia, PA, 19104, USA.
| |
Collapse
|
46
|
Hadas Y, Katz MG, Bridges CR, Zangi L. Modified mRNA as a therapeutic tool to induce cardiac regeneration in ischemic heart disease. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2016; 9. [PMID: 27911047 DOI: 10.1002/wsbm.1367] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 10/03/2016] [Accepted: 10/12/2016] [Indexed: 01/14/2023]
Abstract
Ischemic heart disease (IHD) is a leading cause of morbidity and mortality in developed countries. Current pharmacological and interventional therapies provide significant improvement in the life quality of patient; however, they are mostly symptom-oriented and not curative. A high disease and economic burden of IHD requires the search for new therapeutic strategies to significantly improve patients' prognosis and quality of life. One of the main challenges during IHD is the massive loss of cardiomyocytes that possess minimal regenerative capacity. Recent understanding of the pathophysiological mechanisms underlying IHD, as well as new therapeutic approaches provide new hope for patients suffering from IHD. Synthetic modified mRNA (modRNA) is a new gene delivery vector that is increasingly used in in vivo applications. modRNA is a relatively stable, non-immunogenic, highly-expressed molecule that has been shown to mediate high and transient expression of proteins in different type of cells and tissues including cardiomyocytes. modRNA properties, together with its expression kinetics in the heart make it an attractive option for the treatment of IHD, especially after myocardial infarction. In this review we discuss the role of gene therapy in cardiac regeneration as an approach to treat IHD; traditional and innovative gene delivery methods; and focus specifically on modRNA structure, mode of delivery, and its use for the induction of endogenous regenerative capacity, mainly in the context of IHD. WIREs Syst Biol Med 2017, 9:e1367. doi: 10.1002/wsbm.1367 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Yoav Hadas
- Cardiovascular Research Center, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael G Katz
- Cardiovascular Research Center, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Charles R Bridges
- Cardiovascular Research Center, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lior Zangi
- Cardiovascular Research Center, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
47
|
Reautschnig P, Vogel P, Stafforst T. The notorious R.N.A. in the spotlight - drug or target for the treatment of disease. RNA Biol 2016; 14:651-668. [PMID: 27415589 PMCID: PMC5449091 DOI: 10.1080/15476286.2016.1208323] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
mRNA is an attractive drug target for therapeutic interventions. In this review we highlight the current state, clinical trials, and developments in antisense therapy, including the classical approaches like RNaseH-dependent oligomers, splice-switching oligomers, aptamers, and therapeutic RNA interference. Furthermore, we provide an overview on emerging concepts for using RNA in therapeutic settings including protein replacement by in-vitro-transcribed mRNAs, mRNA as vaccines and anti-allergic drugs. Finally, we give a brief outlook on early-stage RNA repair approaches that apply endogenous or engineered proteins in combination with short RNAs or chemically stabilized oligomers for the re-programming of point mutations, RNA modifications, and frame shift mutations directly on the endogenous mRNA.
Collapse
Affiliation(s)
- Philipp Reautschnig
- a Interfaculty Institute of Biochemistry, University of Tübingen Auf der Morgenstelle , Tübingen , Germany
| | - Paul Vogel
- a Interfaculty Institute of Biochemistry, University of Tübingen Auf der Morgenstelle , Tübingen , Germany
| | - Thorsten Stafforst
- a Interfaculty Institute of Biochemistry, University of Tübingen Auf der Morgenstelle , Tübingen , Germany
| |
Collapse
|
48
|
Abstract
The study of RNA has continually emphasized the structural and functional versatility of RNA molecules. This versatility has inspired translational and clinical researchers to explore the utility of RNA-based therapeutic agents for a wide variety of medical applications. Several RNA therapeutics, with diverse modes of action, are being evaluated in large late-stage clinical trials, and many more are in early clinical development. Hundreds of patients are enrolled in large trials testing messenger RNAs to combat cancer, small interfering RNAs to treat renal and hepatic disorders, and aptamers to combat ocular and cardiovascular disease. Results from these studies are generating considerable interest among the biomedical community and the public and will be important for the future development of this emerging class of therapeutic agents.
Collapse
Affiliation(s)
- Bruce A Sullenger
- Duke Translational Research Institute and Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Smita Nair
- Duke Translational Research Institute and Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
49
|
Lucas CGDO, Matassoli FL, Peçanha LMT, Santillo BT, Oliveira LMDS, Oshiro TM, Marques ETDA, Oxenius A, de Arruda LB. Dendritic cells primed with a chimeric plasmid containing HIV-1-gag associated with lysosomal-associated protein-1 (LAMP/gag) is a potential therapeutic vaccine against HIV. FASEB J 2016; 30:2970-84. [PMID: 27199296 DOI: 10.1096/fj.201500059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 05/02/2016] [Indexed: 11/11/2022]
Abstract
The decline in number and function of T cells is a hallmark of HIV infection, and preservation or restoration of HIV-specific cellular immune response is a major goal of AIDS treatment. Dendritic cells (DCs) play a key role in the initiation and maintenance of the immune response, and their use as a vaccine vehicle is a promising strategy for enhancing vaccine efficacy. We evaluated the potential of DC-mediated immunization with a DNA vaccine consisting of HIV-1-p55gag (gag, group-specific antigen) associated to lysosomal associated protein (LAMP) sequence (LAMP/gag vaccine). Immunization of mice with mouse DCs transfected with LAMP/gag (Lg-mDCs) stimulated more potent B- and T-cell responses than naked DNA or DCs pulsed with inactivated HIV. Anti-Gag antibody levels were sustained for at least 3 mo after immunization, and recall T-cell responses were also strongly detected at this time point. Human DCs transfected with LAMP/gag (Lg-hDCs) were also activated and able to stimulate greater T-cell response than native gag-transfected DCs. Coculture between Lg-hDCs and T lymphocytes obtained from patients with HIV resulted in upregulation of CD38, CD69, HLA-DR, and granzyme B by CD4(+) and CD8(+) T cells, and increased IFN-γ and TNF-α production. These results indicate that the use of LAMP/gag-DC may be an efficient strategy for enhancing immune function in patients with HIV.-Lucas, C. G. D. O., Matassoli, F. L., Peçanha, L. M. T., Santillo, B. T., Oliveira, L. M. D. S., Oshiro, T. M., Marques, E. T. D. A., Jr., Oxenius, A., de Arruda, L. B. Dendritic cells primed with a chimeric plasmid containing HIV-1-gag associated with lysosomal-associated protein-1 (LAMP/gag) is a potential therapeutic vaccine against HIV.
Collapse
Affiliation(s)
- Carolina G D O Lucas
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Flavio L Matassoli
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ligia M T Peçanha
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Bruna Tereso Santillo
- Laboratório de Dermatologia e Imunodeficiências (LIM-56), Departamento de Dermatologia, Escola de Medicina, Universidade de São Paulo, São Paulo, Brazil; Institute of Microbiology, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Luanda Mara da Silva Oliveira
- Laboratório de Dermatologia e Imunodeficiências (LIM-56), Departamento de Dermatologia, Escola de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Telma Miyuki Oshiro
- Laboratório de Dermatologia e Imunodeficiências (LIM-56), Departamento de Dermatologia, Escola de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Ernesto T D A Marques
- Department of Infectious Diseases and Microbiology, Center for Vaccine Research, Pittsburgh, Pennsylvania, USA; and Department of Virology, Centro de Pesquisas Aggeu Magalhães (CPqAM), Fundação Oswaldo Cruz (Fiocruz)-Pernambuco, Recife, Brazil
| | - Annette Oxenius
- Institute of Microbiology, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Luciana B de Arruda
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil;
| |
Collapse
|
50
|
Zhao C, Ao Z, Yao X. Current Advances in Virus-Like Particles as a Vaccination Approach against HIV Infection. Vaccines (Basel) 2016; 4:vaccines4010002. [PMID: 26805898 PMCID: PMC4810054 DOI: 10.3390/vaccines4010002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 12/31/2015] [Accepted: 01/18/2016] [Indexed: 12/16/2022] Open
Abstract
HIV-1 virus-like particles (VLPs) are promising vaccine candidates against HIV-1 infection. They are capable of preserving the native conformation of HIV-1 antigens and priming CD4+ and CD8+ T cell responses efficiently via cross presentation by both major histocompatibility complex (MHC) class I and II molecules. Progress has been achieved in the preclinical research of HIV-1 VLPs as prophylactic vaccines that induce broadly neutralizing antibodies and potent T cell responses. Moreover, the progress in HIV-1 dendritic cells (DC)-based immunotherapy provides us with a new vision for HIV-1 vaccine development. In this review, we describe updates from the past 5 years on the development of HIV-1 VLPs as a vaccine candidate and on the combined use of HIV particles with HIV-1 DC-based immunotherapy as efficient prophylactic and therapeutic vaccination strategies.
Collapse
Affiliation(s)
- Chongbo Zhao
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| | - Zhujun Ao
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| | - Xiaojian Yao
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
- Department of Microbiology, School of Basic Medical Sciences, Central South University, Changsha 410078, Hunan, China.
| |
Collapse
|