1
|
Muthukutty P, MacDonald J, Yoo SY. Combating Emerging Respiratory Viruses: Lessons and Future Antiviral Strategies. Vaccines (Basel) 2024; 12:1220. [PMID: 39591123 PMCID: PMC11598775 DOI: 10.3390/vaccines12111220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/28/2024] Open
Abstract
Emerging viral diseases, including seasonal illnesses and pandemics, pose significant global public health risks. Respiratory viruses, particularly coronaviruses and influenza viruses, are associated with high morbidity and mortality, imposing substantial socioeconomic burdens. This review focuses on the current landscape of respiratory viruses, particularly influenza and SARS-CoV-2, and their antiviral treatments. It also discusses the potential for pandemics and the development of new antiviral vaccines and therapies, drawing lessons from past outbreaks to inform future strategies for managing viral threats.
Collapse
Affiliation(s)
| | | | - So Young Yoo
- Institute of Nanobio Convergence, Pusan National University, Busan 46241, Republic of Korea; (P.M.); (J.M.)
| |
Collapse
|
2
|
Oelkrug C. Analysis of physical and biological delivery systems for DNA cancer vaccines and their translation to clinical development. Clin Exp Vaccine Res 2024; 13:73-82. [PMID: 38752006 PMCID: PMC11091436 DOI: 10.7774/cevr.2024.13.2.73] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/17/2023] [Accepted: 03/30/2024] [Indexed: 05/18/2024] Open
Abstract
DNA cancer vaccines as an approach in tumor immunotherapy are still being investigated in preclinical and clinical settings. Nevertheless, only a small number of clinical studies have been published so far and are still active. The investigated vaccines show a relatively stable expression in in-vitro transfected cells and may be favorable for developing an immunologic memory in patients. Therefore, DNA vaccines could be suitable as a prophylactic or therapeutic approach against cancer. Due to the low efficiency of these vaccines, the administration technique plays an important role in the vaccine design and its efficacy. These DNA cancer vaccine delivery systems include physical, biological, and non-biological techniques. Although the pre-clinical studies show promising results in the application of the different delivery systems, further studies in clinical trials have not yet been successfully proven.
Collapse
|
3
|
Bostanghadiri N, Ziaeefar P, Mofrad MG, Yousefzadeh P, Hashemi A, Darban-Sarokhalil D. COVID-19: An Overview of SARS-CoV-2 Variants-The Current Vaccines and Drug Development. BIOMED RESEARCH INTERNATIONAL 2023; 2023:1879554. [PMID: 37674935 PMCID: PMC10480030 DOI: 10.1155/2023/1879554] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 07/07/2023] [Accepted: 08/04/2023] [Indexed: 09/08/2023]
Abstract
The world is presently in crisis facing an outbreak of a health-threatening microorganism known as COVID-19, responsible for causing uncommon viral pneumonia in humans. The virus was first reported in Wuhan, China, in early December 2019, and it quickly became a global concern due to the pandemic. Challenges in this regard have been compounded by the emergence of several variants such as B.1.1.7, B.1.351, P1, and B.1.617, which show an increase in transmission power and resistance to therapies and vaccines. Ongoing researches are focused on developing and manufacturing standard treatment strategies and effective vaccines to control the pandemic. Despite developing several vaccines such as Pfizer/BioNTech and Moderna approved by the U.S. Food and Drug Administration (FDA) and other vaccines in phase 4 clinical trials, preventive measures are mandatory to control the COVID-19 pandemic. In this review, based on the latest findings, we will discuss different types of drugs as therapeutic options and confirmed or developing vaccine candidates against SARS-CoV-2. We also discuss in detail the challenges posed by the variants and their effect on therapeutic and preventive interventions.
Collapse
Affiliation(s)
- Narjess Bostanghadiri
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Pardis Ziaeefar
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Morvarid Golrokh Mofrad
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Parsa Yousefzadeh
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Hashemi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Darban-Sarokhalil
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Saeed Y. Title: Immunotherapy; a ground-breaking remedy for spinal cord injury with stumbling blocks: An overview. Front Pharmacol 2023; 14:1110008. [PMID: 36778022 PMCID: PMC9909832 DOI: 10.3389/fphar.2023.1110008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 01/11/2023] [Indexed: 01/26/2023] Open
Abstract
Spinal cord injury (SCI) is a debilitating disorder with no known standard and effective treatment. Despite its ability to exacerbate SCI sequel by accelerating auto-reactive immune cells, an immune response is also considered essential to the healing process. Therefore, immunotherapeutic strategies targeting spinal cord injuries may benefit from the dual nature of immune responses. An increasing body of research suggests that immunization against myelin inhibitors can promote axon remyelination after SCI. However, despite advancements in our understanding of neuroimmune responses, immunoregulation-based therapeutic strategies have yet to receive widespread acceptance. Therefore, it is a prerequisite to enhance the understanding of immune regulation to ensure the safety and efficacy of immunotherapeutic treatments. The objective of the present study was to provide an overview of previous studies regarding the advantages and limitations of immunotherapeutic strategies for functional recovery after spinal cord injury, especially in light of limiting factors related to DNA and cell-based vaccination strategies by providing a novel prospect to lay the foundation for future studies that will help devise a safe and effective treatment for spinal cord injury.
Collapse
Affiliation(s)
- Yasmeen Saeed
- Provincial Key Laboratory for Utilization and Conservation of Food and Medicinal Resources in Northern Guangdong, 288 University Ave. Zhenjiang District, Shaoguan City, Guangdong Province, China
| |
Collapse
|
5
|
Ramos RN, Tosch C, Kotsias F, Claudepierre MC, Schmitt D, Remy-Ziller C, Hoffmann C, Ricordel M, Nourtier V, Farine I, Laruelle L, Hortelano J, Spring-Giusti C, Sedlik C, Le Tourneau C, Hoffmann C, Silvestre N, Erbs P, Bendjama K, Thioudellet C, Quemeneur E, Piaggio E, Rittner K. Pseudocowpox virus, a novel vector to enhance the therapeutic efficacy of antitumor vaccination. Clin Transl Immunology 2022; 11:e1392. [PMID: 35573979 PMCID: PMC9081486 DOI: 10.1002/cti2.1392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 01/11/2022] [Accepted: 04/16/2022] [Indexed: 11/11/2022] Open
Abstract
Objective Antitumor viral vaccines, and more particularly poxviral vaccines, represent an active field for clinical development and translational research. To improve the efficacy and treatment outcome, new viral vectors are sought, with emphasis on their abilities to stimulate innate immunity, to display tumor antigens and to induce a specific T‐cell response. Methods We screened for a new poxviral backbone with improved innate and adaptive immune stimulation using IFN‐α secretion levels in infected PBMC cultures as selection criteria. Assessment of virus effectiveness was made in vitro and in vivo. Results The bovine pseudocowpox virus (PCPV) stood out among several poxviruses for its ability to induce significant secretion of IFN‐α. PCPV produced efficient activation of human monocytes and dendritic cells, degranulation of NK cells and reversed MDSC‐induced T‐cell suppression, without being offensive to activated T cells. A PCPV‐based vaccine, encoding the HPV16 E7 protein (PCPV‐E7), stimulated strong antigen‐specific T‐cell responses in TC1 tumor‐bearing mice. Complete regression of tumors was obtained in a CD8+ T‐cell‐dependent manner after intratumoral injection of PCPV‐E7, followed by intravenous injection of the cancer vaccine MVA‐E7. PCPV also proved active when injected repeatedly intratumorally in MC38 tumor‐bearing mice, generating tumor‐specific T‐cell responses without encoding a specific MC38 antigen. From a translational perspective, we demonstrated that PCPV‐E7 effectively stimulated IFN‐γ production by T cells from tumor‐draining lymph nodes of HPV+‐infected cancer patients. Conclusion We propose PCPV as a viral vector suitable for vaccination in the field of personalised cancer vaccines, in particular for heterologous prime‐boost regimens.
Collapse
Affiliation(s)
- Rodrigo Nalio Ramos
- Institut Curie INSERM U932, and Centre d'Investigation Clinique Biotherapie CICBT 1428 PSL Research University Paris France.,Present address: Laboratório de Investigação Médica em Patogênese e Terapia dirigida em Onco-Imuno-Hematologia Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo (HCFMUSP) São Paulo Brazil.,Present address: Instituto D'Or de Ensino e Pesquisa São Paulo Brazil
| | | | - Fiorella Kotsias
- Institut Curie INSERM U932, and Centre d'Investigation Clinique Biotherapie CICBT 1428 PSL Research University Paris France
| | | | | | | | | | | | | | | | | | | | | | - Christine Sedlik
- Institut Curie INSERM U932, and Centre d'Investigation Clinique Biotherapie CICBT 1428 PSL Research University Paris France
| | - Christophe Le Tourneau
- Department of Drug Development and Innovation (D3i) Institut Curie Paris and Saint-Cloud France
| | - Caroline Hoffmann
- Institut Curie INSERM U932, and Centre d'Investigation Clinique Biotherapie CICBT 1428 PSL Research University Paris France.,Department of Surgical Oncology Institut Curie PSL Research University Paris France
| | | | | | | | | | | | - Eliane Piaggio
- Institut Curie INSERM U932, and Centre d'Investigation Clinique Biotherapie CICBT 1428 PSL Research University Paris France
| | | |
Collapse
|
6
|
Mosharaf MP, Reza MS, Gov E, Mahumud RA, Mollah MNH. Disclosing Potential Key Genes, Therapeutic Targets and Agents for Non-Small Cell Lung Cancer: Evidence from Integrative Bioinformatics Analysis. Vaccines (Basel) 2022; 10:vaccines10050771. [PMID: 35632527 PMCID: PMC9143695 DOI: 10.3390/vaccines10050771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 05/07/2022] [Accepted: 05/08/2022] [Indexed: 12/10/2022] Open
Abstract
Non-small-cell lung cancer (NSCLC) is considered as one of the malignant cancers that causes premature death. The present study aimed to identify a few potential novel genes highlighting their functions, pathways, and regulators for diagnosis, prognosis, and therapies of NSCLC by using the integrated bioinformatics approaches. At first, we picked out 1943 DEGs between NSCLC and control samples by using the statistical LIMMA approach. Then we selected 11 DEGs (CDK1, EGFR, FYN, UBC, MYC, CCNB1, FOS, RHOB, CDC6, CDC20, and CHEK1) as the hub-DEGs (potential key genes) by the protein–protein interaction network analysis of DEGs. The DEGs and hub-DEGs regulatory network analysis commonly revealed four transcription factors (FOXC1, GATA2, YY1, and NFIC) and five miRNAs (miR-335-5p, miR-26b-5p, miR-92a-3p, miR-155-5p, and miR-16-5p) as the key transcriptional and post-transcriptional regulators of DEGs as well as hub-DEGs. We also disclosed the pathogenetic processes of NSCLC by investigating the biological processes, molecular function, cellular components, and KEGG pathways of DEGs. The multivariate survival probability curves based on the expression of hub-DEGs in the SurvExpress web-tool and database showed the significant differences between the low- and high-risk groups, which indicates strong prognostic power of hub-DEGs. Then, we explored top-ranked 5-hub-DEGs-guided repurposable drugs based on the Connectivity Map (CMap) database. Out of the selected drugs, we validated six FDA-approved launched drugs (Dinaciclib, Afatinib, Icotinib, Bosutinib, Dasatinib, and TWS-119) by molecular docking interaction analysis with the respective target proteins for the treatment against NSCLC. The detected therapeutic targets and repurposable drugs require further attention by experimental studies to establish them as potential biomarkers for precision medicine in NSCLC treatment.
Collapse
Affiliation(s)
- Md. Parvez Mosharaf
- Bioinformatics Lab, Department of Statistics, University of Rajshahi, Rajshahi 6205, Bangladesh; (M.P.M.); (M.S.R.)
- School of Commerce, Faculty of Business, Education, Law and Arts, University of Southern Queensland, Toowoomba, QLD 4350, Australia
| | - Md. Selim Reza
- Bioinformatics Lab, Department of Statistics, University of Rajshahi, Rajshahi 6205, Bangladesh; (M.P.M.); (M.S.R.)
- Centre for High Performance Computing, Joint Engineering Research Centre for Health Big Data Intelligent Analysis Technology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Esra Gov
- Department of Bioengineering, Faculty of Engineering, Adana AlparslanTurkes Science and Technology University, Adana 01250, Turkey;
| | - Rashidul Alam Mahumud
- NHMRC Clinical Trials Centre, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia;
| | - Md. Nurul Haque Mollah
- Bioinformatics Lab, Department of Statistics, University of Rajshahi, Rajshahi 6205, Bangladesh; (M.P.M.); (M.S.R.)
- Correspondence:
| |
Collapse
|
7
|
Sun B, Zhao X, Gu W, Cao P, Movahedi F, Wu Y, Xu ZP, Gu W. ATP stabilised and sensitised calcium phosphate nanoparticles as effective adjuvants for a DNA vaccine against cancer. J Mater Chem B 2021; 9:7435-7446. [PMID: 34551058 DOI: 10.1039/d1tb01408k] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cancer vaccines based on DNA encoding oncogenes have shown great potential in preclinical studies. However, the efficacy of DNA vaccines is limited by their weak immunogenicity because of low cellular internalisation and insufficient activation of dendritic cells (DCs). Calcium phosphate (CP) nanoparticles (NPs) are biodegradable vehicles with low toxicity and high loading capacity of DNA but suffer from stability issues. Here we employed adenosine triphosphate (ATP) as a dual functional agent, i.e. stabiliser for CP and immunological adjuvant, and applied the ATP-modified CP (ACP) NPs to the DNA vaccine. ACP NP-enhanced cellular uptake and improved transfection efficiency of DNA vaccine, and further showed the ability to activate DCs that are critical for them to prime T cells in cancer immunotherapy. As a result, a higher level of antigen-specific antibody with stronger tumour growth inhibition was achieved in mice immunised with the ACP-DNA vaccine. Overall, this one-step synthesised ACP NPs are an efficient nano-delivery system and nano-adjuvant for cancer DNA vaccines.
Collapse
Affiliation(s)
- Bing Sun
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Building 75, Corner of Cooper Road & College Road, St Lucia, QLD 4072, Australia.
| | - Xiaohui Zhao
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Building 75, Corner of Cooper Road & College Road, St Lucia, QLD 4072, Australia. .,GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Wenxi Gu
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Building 75, Corner of Cooper Road & College Road, St Lucia, QLD 4072, Australia. .,Institute of Veterinary Medicine, Xinjiang Academy of Animal Science, Urumqi, 830011, China
| | - Pei Cao
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Building 75, Corner of Cooper Road & College Road, St Lucia, QLD 4072, Australia.
| | - Fatemeh Movahedi
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Building 75, Corner of Cooper Road & College Road, St Lucia, QLD 4072, Australia.
| | - Yanheng Wu
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Building 75, Corner of Cooper Road & College Road, St Lucia, QLD 4072, Australia. .,Gillion ITM Research Institute, Guangzhou Hongkeyuan, Guangzhou, 510530, China
| | - Zhi Ping Xu
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Building 75, Corner of Cooper Road & College Road, St Lucia, QLD 4072, Australia.
| | - Wenyi Gu
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Building 75, Corner of Cooper Road & College Road, St Lucia, QLD 4072, Australia. .,Gillion ITM Research Institute, Guangzhou Hongkeyuan, Guangzhou, 510530, China
| |
Collapse
|
8
|
Chong WC, Chellappan DK, Shukla SD, Peterson GM, Patel RP, Jha NK, Eri RD, Dua K, Tambuwala MM, Shastri MD. An Appraisal of the Current Scenario in Vaccine Research for COVID-19. Viruses 2021; 13:1397. [PMID: 34372603 PMCID: PMC8310376 DOI: 10.3390/v13071397] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 12/15/2022] Open
Abstract
The recent coronavirus disease 2019 (COVID-19) outbreak has drawn global attention, affecting millions, disrupting economies and healthcare modalities. With its high infection rate, COVID-19 has caused a colossal health crisis worldwide. While information on the comprehensive nature of this infectious agent, SARS-CoV-2, still remains obscure, ongoing genomic studies have been successful in identifying its genomic sequence and the presenting antigen. These may serve as promising, potential therapeutic targets in the effective management of COVID-19. In an attempt to establish herd immunity, massive efforts have been directed and driven toward developing vaccines against the SARS-CoV-2 pathogen. This review, in this direction, is aimed at providing the current scenario and future perspectives in the development of vaccines against SARS-CoV-2.
Collapse
Affiliation(s)
- Wai Chin Chong
- Department of Molecular and Translational Science, Monash University, Clayton 3168, Australia;
| | - Dinesh K. Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University (IMU), Kuala Lumpur 57000, Malaysia;
| | - Shakti D. Shukla
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney 2007, Australia; (S.D.S.); (K.D.)
| | - Gregory M. Peterson
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart 7005, Australia; (G.M.P.); (R.P.P.)
| | - Rahul P. Patel
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart 7005, Australia; (G.M.P.); (R.P.P.)
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida 201310, UP, India;
| | - Rajaraman D. Eri
- School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston 7248, Australia;
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney 2007, Australia; (S.D.S.); (K.D.)
| | - Murtaza M. Tambuwala
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine BT52 1SA, UK
| | - Madhur D. Shastri
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart 7005, Australia; (G.M.P.); (R.P.P.)
- School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston 7248, Australia;
| |
Collapse
|
9
|
Varadé J, Magadán S, González-Fernández Á. Human immunology and immunotherapy: main achievements and challenges. Cell Mol Immunol 2021; 18:805-828. [PMID: 32879472 PMCID: PMC7463107 DOI: 10.1038/s41423-020-00530-6] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/27/2020] [Accepted: 07/31/2020] [Indexed: 02/07/2023] Open
Abstract
The immune system is a fascinating world of cells, soluble factors, interacting cells, and tissues, all of which are interconnected. The highly complex nature of the immune system makes it difficult to view it as a whole, but researchers are now trying to put all the pieces of the puzzle together to obtain a more complete picture. The development of new specialized equipment and immunological techniques, genetic approaches, animal models, and a long list of monoclonal antibodies, among many other factors, are improving our knowledge of this sophisticated system. The different types of cell subsets, soluble factors, membrane molecules, and cell functionalities are some aspects that we are starting to understand, together with their roles in health, aging, and illness. This knowledge is filling many of the gaps, and in some cases, it has led to changes in our previous assumptions; e.g., adaptive immune cells were previously thought to be unique memory cells until trained innate immunity was observed, and several innate immune cells with features similar to those of cytokine-secreting T cells have been discovered. Moreover, we have improved our knowledge not only regarding immune-mediated illnesses and how the immune system works and interacts with other systems and components (such as the microbiome) but also in terms of ways to manipulate this system through immunotherapy. The development of different types of immunotherapies, including vaccines (prophylactic and therapeutic), and the use of pathogens, monoclonal antibodies, recombinant proteins, cytokines, and cellular immunotherapies, are changing the way in which we approach many diseases, especially cancer.
Collapse
Affiliation(s)
- Jezabel Varadé
- CINBIO, Centro de Investigaciones Biomédicas, Universidade de Vigo, Immunology Group, Campus Universitario Lagoas, Marcosende, 36310, Vigo, Spain
- Instituto de Investigación Sanitaria Galicia Sur (IIS-Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Susana Magadán
- CINBIO, Centro de Investigaciones Biomédicas, Universidade de Vigo, Immunology Group, Campus Universitario Lagoas, Marcosende, 36310, Vigo, Spain
- Instituto de Investigación Sanitaria Galicia Sur (IIS-Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - África González-Fernández
- CINBIO, Centro de Investigaciones Biomédicas, Universidade de Vigo, Immunology Group, Campus Universitario Lagoas, Marcosende, 36310, Vigo, Spain.
- Instituto de Investigación Sanitaria Galicia Sur (IIS-Galicia Sur), SERGAS-UVIGO, Vigo, Spain.
| |
Collapse
|
10
|
Sun B, Zhao X, Wu Y, Cao P, Movahedi F, Liu J, Wang J, Xu ZP, Gu W. Mannose-Functionalized Biodegradable Nanoparticles Efficiently Deliver DNA Vaccine and Promote Anti-tumor Immunity. ACS APPLIED MATERIALS & INTERFACES 2021; 13:14015-14027. [PMID: 33751882 DOI: 10.1021/acsami.1c01401] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Cancer vaccines have attracted increasing attention for their application in tumor immunotherapy. DNA vaccines are one of them that have been proven very promising with the advantages of safety, rapid design, and low cost. However, the low stability, ineffective cell internalization, and low immunostimulation hinder their wide application. Thus, developing targeted and safe systems to effectively deliver DNA vaccines becomes a vital step. In this study, we report the development of mannose- and bisphosphonate (BP)-modified calcium phosphate (CP) nanoparticles (NPs) as efficient vaccine delivery vehicles by targeting C-type lectin receptors (CLRs) on antigen-presenting cells (APCs). Using a model antigen ovalbumin (OVA)-encoded plasmid DNA (pOVA) as a model vaccine, we demonstrate that mannose-modified and BP-stabilized CP (MBCP) nanoparticles are mono-dispersed for enhanced uptake by APCs and subsequently induce OVA antigen presentation and immunostimulation. Mice immunized with MBCP-pOVA nanovaccines show a significantly stronger anti-OVA antibody response with a quicker IgG1 and IgG2a antibody production than unmodified NPs. Moreover, MBCP-pOVA immunization significantly inhibits the growth of OVA-expressing E.G7 tumor cells in C57BL/6J mice. Our data collectively suggest that the modifications to enhance the stability and targeting ability of MBCP NPs are essential for effective delivery of DNA vaccines and promote robust anti-tumor immunity.
Collapse
Affiliation(s)
- Bing Sun
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Xiaohui Zhao
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland 4072, Australia
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Yanheng Wu
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland 4072, Australia
- Gillion ITM Research Institute, Guangzhou Hongkeyuan, Guangzhou 510530, China
| | - Pei Cao
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Fatemeh Movahedi
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Jie Liu
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Jingjing Wang
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Zhi Ping Xu
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Wenyi Gu
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland 4072, Australia
| |
Collapse
|
11
|
Zawawi A, Else KJ. Soil-Transmitted Helminth Vaccines: Are We Getting Closer? Front Immunol 2020; 11:576748. [PMID: 33133094 PMCID: PMC7565266 DOI: 10.3389/fimmu.2020.576748] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/02/2020] [Indexed: 01/07/2023] Open
Abstract
Parasitic helminths infect over one-fourth of the human population resulting in significant morbidity, and in some cases, death in endemic countries. Despite mass drug administration (MDA) to school-aged children and other control measures, helminth infections are spreading into new areas. Thus, there is a strong rationale for developing anthelminthic vaccines as cost-effective, long-term immunological control strategies, which, unlike MDA, are not haunted by the threat of emerging drug-resistant helminths nor limited by reinfection risk. Advances in vaccinology, immunology, and immunomics include the development of new tools that improve the safety, immunogenicity, and efficacy of vaccines; and some of these tools have been used in the development of helminth vaccines. The development of anthelminthic vaccines is fraught with difficulty. Multiple lifecycle stages exist each presenting stage-specific antigens. Further, helminth parasites are notorious for their ability to dampen down and regulate host immunity. One of the first significant challenges in developing any vaccine is identifying suitable candidate protective antigens. This review explores our current knowledge in lead antigen identification and reports on recent pre-clinical and clinical trials in the context of the soil-transmitted helminths Trichuris, the hookworms and Ascaris. Ultimately, a multivalent anthelminthic vaccine could become an essential tool for achieving the medium-to long-term goal of controlling, or even eliminating helminth infections.
Collapse
Affiliation(s)
- Ayat Zawawi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia,*Correspondence: Ayat Zawawi
| | - Kathryn J. Else
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine, and Health, School of Biological Sciences, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom,Kathryn J. Else
| |
Collapse
|
12
|
Adam L, Tchitchek N, Todorova B, Rosenbaum P, Joly C, Poux C, Chapon C, Spetz AL, Ustav M, Le Grand R, Martinon F. Innate Molecular and Cellular Signature in the Skin Preceding Long-Lasting T Cell Responses after Electroporated DNA Vaccination. THE JOURNAL OF IMMUNOLOGY 2020; 204:3375-3388. [PMID: 32385135 DOI: 10.4049/jimmunol.1900517] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 04/09/2020] [Indexed: 12/21/2022]
Abstract
DNA vaccines delivered with electroporation (EP) have shown promising results in preclinical models and are evaluated in clinical trials. In this study, we aim to characterize early mechanisms occurring in the skin after intradermal injection and EP of the auxoGTUmultiSIV DNA vaccine in nonhuman primates. First, we show that EP acts as an adjuvant by enhancing local inflammation, notably via granulocytes, monocytes/macrophages, and CD1aint-expressing cell recruitment. EP also induced Langerhans cell maturation, illustrated by CD86, CD83, and HLA-DR upregulation and their migration out of the epidermis. Second, we demonstrate the crucial role of the DNA vaccine in soluble factors release, such as MCP-1 or IL-15. Transcriptomic analysis showed that EP played a major role in gene expression changes postvaccination. However, the DNA vaccine is required to strongly upregulate several genes involved in inflammatory responses (e.g., Saa4), cell migration (e.g., Ccl3, Ccl5, or Cxcl10), APC activation (e.g., Cd86), and IFN-inducible genes (e.g., Ifit3, Ifit5, Irf7, Isg15, orMx1), illustrating an antiviral response signature. Also, AIM-2, a cytosolic DNA sensor, appeared to be strongly upregulated only in the presence of the DNA vaccine and trends to positively correlate with several IFN-inducible genes, suggesting the potential role of AIM-2 in vaccine sensing and the subsequent innate response activation leading to strong adaptive T cell responses. Overall, these results demonstrate that a combined stimulation of the immune response, in which EP and the auxoGTUmultiSIV vaccine triggered different components of the innate immunity, led to strong and persistent cellular recall responses.
Collapse
Affiliation(s)
- Lucille Adam
- Immunology of Viral Infections and Autoimmune Diseases, Infectious Disease Models and Innovative Therapies Department, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Paris-Sud 11, INSERM U1184, 92265 Fontenay-aux-Roses, France
| | - Nicolas Tchitchek
- Immunology of Viral Infections and Autoimmune Diseases, Infectious Disease Models and Innovative Therapies Department, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Paris-Sud 11, INSERM U1184, 92265 Fontenay-aux-Roses, France
| | - Biliana Todorova
- Immunology of Viral Infections and Autoimmune Diseases, Infectious Disease Models and Innovative Therapies Department, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Paris-Sud 11, INSERM U1184, 92265 Fontenay-aux-Roses, France
| | - Pierre Rosenbaum
- Immunology of Viral Infections and Autoimmune Diseases, Infectious Disease Models and Innovative Therapies Department, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Paris-Sud 11, INSERM U1184, 92265 Fontenay-aux-Roses, France
| | - Candie Joly
- Immunology of Viral Infections and Autoimmune Diseases, Infectious Disease Models and Innovative Therapies Department, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Paris-Sud 11, INSERM U1184, 92265 Fontenay-aux-Roses, France
| | - Candice Poux
- Immunology of Viral Infections and Autoimmune Diseases, Infectious Disease Models and Innovative Therapies Department, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Paris-Sud 11, INSERM U1184, 92265 Fontenay-aux-Roses, France
| | - Catherine Chapon
- Immunology of Viral Infections and Autoimmune Diseases, Infectious Disease Models and Innovative Therapies Department, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Paris-Sud 11, INSERM U1184, 92265 Fontenay-aux-Roses, France
| | - Anna-Lena Spetz
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden; and
| | - Mart Ustav
- Institute of Technology, University of Tartu, 50411 Tartu, Estonia
| | - Roger Le Grand
- Immunology of Viral Infections and Autoimmune Diseases, Infectious Disease Models and Innovative Therapies Department, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Paris-Sud 11, INSERM U1184, 92265 Fontenay-aux-Roses, France
| | - Frédéric Martinon
- Immunology of Viral Infections and Autoimmune Diseases, Infectious Disease Models and Innovative Therapies Department, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Paris-Sud 11, INSERM U1184, 92265 Fontenay-aux-Roses, France;
| |
Collapse
|
13
|
Neukirch L, Fougeroux C, Andersson AMC, Holst PJ. The potential of adenoviral vaccine vectors with altered antigen presentation capabilities. Expert Rev Vaccines 2020; 19:25-41. [PMID: 31889453 DOI: 10.1080/14760584.2020.1711054] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Despite their appeal as vaccine vectors, adenoviral vectors are yet unable to induce protective immune responses against some weakly immunogenic antigens. Additionally, the maximum doses of adenovirus-based vaccines are limited by vector-induced toxicity, causing vector elimination and diminished immune responses against the target antigen. In order to increase immune responses to the transgene, while maintaining a moderate vector dose, new technologies for improved transgene presentation have been developed for adenoviral vaccine vectors.Areas covered: This review provides an overview of different genetic-fusion adjuvants that aim to improve antigen presentation in the context of adenoviral vector-based vaccines. The influence on both T cell and B cell responses are discussed, with a main focus on two technologies: MHC class II-associated invariant chain and virus-like-vaccines.Expert opinion: Different strategies have been tested to improve adenovirus-based vaccinations with varying degrees of success. The reviewed genetic adjuvants were designed to increase antigen processing and MHC presentation, or promote humoral immune responses with an improved conformational antigen display. While none of the introduced technologies is universally applicable, this review shall give an overview to identify potential improvements for future vaccination approaches.
Collapse
Affiliation(s)
- Lasse Neukirch
- Clinical Cooperation Unit "Applied Tumor Immunity", National Center for Tumor Diseases and German Cancer Research Center, Heidelberg, Germany.,Center for Medical Parasitology, Department of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Cyrielle Fougeroux
- Center for Medical Parasitology, Department of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Anne-Marie Carola Andersson
- Center for Medical Parasitology, Department of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.,InProTher ApS, Copenhagen, Denmark
| | - Peter Johannes Holst
- Center for Medical Parasitology, Department of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.,InProTher ApS, Copenhagen, Denmark
| |
Collapse
|
14
|
Fernandez MC, Giacani L. Molecular and Immunological Strategies Against Treponema pallidum Infections. Sex Transm Infect 2020. [DOI: 10.1007/978-3-030-02200-6_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
15
|
Brentville VA, Atabani S, Cook K, Durrant LG. Novel tumour antigens and the development of optimal vaccine design. Ther Adv Vaccines Immunother 2018; 6:31-47. [PMID: 29998219 DOI: 10.1177/2515135518768769] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 02/23/2018] [Indexed: 12/13/2022] Open
Abstract
The interplay between tumours and the immune system has long been known to involve complex interactions between tumour cells, immune cells and the tumour microenvironment. The progress of checkpoint inhibitors in the clinic in the last decade has highlighted again the role of the immune system in the fight against cancer. Numerous efforts have been undertaken to develop ways of stimulating the cellular immune response to eradicate tumours. These interventions include the identification of appropriate tumour antigens as targets for therapy. In this review, we summarize progress in selection of target tumour antigen. Targeting self antigens has the problem of thymic deletion of high-affinity T-cell responses leaving a diminished repertoire of low-affinity T cells that fail to kill tumour cells. Thymic regulation appears to be less stringent for differentiation of cancer-testis antigens, as many tumour rejection antigens fall into this category. More recently, targeting neo-epitopes or post-translational modifications such as a phosphorylation or stress-induced citrullination has shown great promise in preclinical studies. Of particular interest is that the responses can be mediated by both CD4 and CD8 T cells. Previous vaccines have targeted CD8 T-cell responses but more recently, the central role of CD4 T cells in orchestrating inflammation within tumours and also differentiating into potent killer cells has been recognized. The design of vaccines to induce such immune responses is discussed herein. Liposomally encoded ribonucleic acid (RNA), targeted deoxyribonucleic acid (DNA) or long peptides linked to toll-like receptor (TLR) adjuvants are the most promising new vaccine approaches. These exciting new approaches suggest that the 'Holy Grail' of a simple nontoxic cancer vaccine may be on the horizon. A major hurdle in tumour therapy is also to overcome the suppressive tumour environment. We address current progress in combination therapies and suggest that these are likely to show the most promise for the future.
Collapse
Affiliation(s)
| | - Suha Atabani
- Academic Department of Clinical Oncology, University of Nottingham, Nottingham, UK
| | - Katherine Cook
- Academic Department of Clinical Oncology, University of Nottingham, Nottingham, UK
| | - Lindy G Durrant
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, City Hospital, Hucknall Road, Nottingham, NG5 1PB, UK
| |
Collapse
|
16
|
Abstract
INTRODUCTION Traditional inactivated and protein vaccines generate strong antibodies, but struggle to generate T cell responses. Attenuated pathogen vaccines generate both, but risk causing the disease they aim to prevent. Newer gene-based vaccines drive both responses and avoid the risk of infection. While these replication-defective (RD) vaccines work well in small animals, they can be weak in humans because they do not replicate antigen genes like more potent replication-competent (RC) vaccines. RC vaccines generate substantially stronger immune responses, but also risk causing their own infections. To circumvent these problems, we developed single-cycle adenovirus (SC-Ad) vectors that amplify vaccine genes, but that avoid the risk of infection. This review will discuss these vectors and their prospects for use as vaccines. AREAS COVERED This review provides a background of different types of vaccines. The benefits of gene-based vaccines and their ability to replicate antigen genes are described. Adenovirus vectors are discussed and compared to other vaccine types. Replication-defective, single-cycle, and replication-competent Ad vaccines are compared. EXPERT COMMENTARY The potential utility of these vaccines are discussed when used against infectious diseases and as cancer vaccines. We propose a move away from replication-defective vaccines towards more robust replication-competent or single-cycle vaccines.
Collapse
Affiliation(s)
- Michael Barry
- a Division of Infectious Diseases, Department of Medicine, Department of Immunology, Department of Molecular Medicine , Mayo Clinic , Rochester , MN , USA
| |
Collapse
|
17
|
A Built-In CpG Adjuvant in RSV F Protein DNA Vaccine Drives a Th1 Polarized and Enhanced Protective Immune Response. Viruses 2018; 10:v10010038. [PMID: 29342954 PMCID: PMC5795451 DOI: 10.3390/v10010038] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 01/08/2018] [Accepted: 01/09/2018] [Indexed: 01/03/2023] Open
Abstract
Human respiratory syncytial virus (RSV) is the most significant cause of acute lower respiratory infection in children. However, there is no licensed vaccine available. Here, we investigated the effect of five or 20 copies of C-Class of CpG ODN (CpG-C) motif incorporated into a plasmid DNA vaccine encoding RSV fusion (F) glycoprotein on the vaccine-induced immune response. The addition of CpG-C motif enhanced serum binding and virus-neutralizing antibody responses in BALB/c mice immunized with the DNA vaccines. Moreover, mice vaccinated with CpG-modified vaccines, especially with the higher 20 copies, resulted in an enhanced shift toward a Th1-biased antibody and T-cell response, a decrease in pulmonary pathology and virus replication, and a decrease in weight loss after RSV challenge. This study suggests that CpG-C motif, cloned into the backbone of DNA vaccine encoding RSV F glycoprotein, functions as a built-in adjuvant capable of improving the efficacy of DNA vaccine against RSV infection.
Collapse
|
18
|
|
19
|
Martins YA, Tsuchida CJ, Antoniassi P, Demarchi IG. Efficacy and Safety of the Immunization with DNA for Alzheimer's Disease in Animal Models: A Systematic Review from Literature. J Alzheimers Dis Rep 2017; 1:195-217. [PMID: 30480238 PMCID: PMC6159633 DOI: 10.3233/adr-170025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disease that does not have a proven cure; however, one of the most promising strategies for its treatment has been DNA vaccines. OBJECTIVE The present review is aimed to report the new developments of the efficacy and safety of DNA vaccines for AD in animal models. METHOD The method PRISMA was used for this review. The article search was made in the electronic databases PubMed, LILACS, and Scopus using the descriptors ''Alzheimer disease" and ''Vaccine, DNA". Articles published between January 2001 and September 2017 in English, Portuguese, and Spanish were included. RESULTS Upon the consensus, the researchers identified 28 original articles. The studies showed satisfying results as for the decrease of amyloid plaques in mouse, rabbits, and monkeys brains using mostly the DNA Aβ42 vaccine, AV-1955, and AdPEDI-(Aβ1-6)11, mainly with a gene gun. In addition to a reduction in tau by the first DNA vaccine (AV-1980D) targeting this protein. The use of adjuvants and boosters also had positive results as they increased the destruction of the amyloid plaques and induced an anti-inflammatory response profile without side effects. CONCLUSION The results of DNA vaccines targeting the amyloid-β and the tau protein with or without adjuvants and boosters were promising in reducing amyloid plaques and tau protein without side effects in animals. Although there are many vaccines being tested in animals, few reach clinical trials. Thus, as a future perspective, we suggest that clinical studies should be conducted with vaccines that have been promising in animal models (e.g., DNA Aβ42 vaccine, AV-1955, and AdPEDI-(Aβ1-6)11).
Collapse
|
20
|
Comparative functional potency of DNA vaccines encoding Plasmodium falciparum transmission blocking target antigens Pfs48/45 and Pfs25 administered alone or in combination by in vivo electroporation in rhesus macaques. Vaccine 2017; 35:7049-7056. [PMID: 29132995 DOI: 10.1016/j.vaccine.2017.10.042] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 09/26/2017] [Accepted: 10/13/2017] [Indexed: 02/07/2023]
Abstract
Antibodies recognizing conformational epitopes in Pfs48/45, an antigen expressed on the surface of Plasmodium falciparum gametes and zygotes, have firmly established Pfs48/45 as a promising transmission blocking vaccine (TBV) candidate. However, it has been difficult to reproducibly express Pfs48/45 in a variety of recombinant expression systems. The goal of our studies was to evaluate functional immunogenicity of Pfs48/45 using DNA vaccine format in rhesus macaques. An additional goal was to ensure that when used in combination with another malarial antigen, specific immunity to both antigens was not compromised. For testing combination vaccines, we employed Pfs25 DNA plasmids that have previously undergone evaluations in rodents and nonhuman primates. Pfs25 is expressed on the surface of parasites after fertilization and is also a lead TBV candidate. DNA plasmids based on codon-optimized sequences of Pfs48/45 and Pfs25 were administered by in vivo electroporation, followed by a final recombinant protein boost. Our studies demonstrate that Pfs48/45 encoded by DNA plasmids is capable of inducing potent transmission blocking antibody responses, and such transmission blocking immune potency of Pfs48/45 was not compromised when tested in combination with Pfs25, These findings provide the evidence in favor of further studies on Pfs48/45 and Pfs25, either alone or in combination with other known malaria vaccine candidates for developing effective vaccines capable of interrupting malaria transmission.
Collapse
|
21
|
Hinkula J, Petkov S, Ljungberg K, Hallengärd D, Bråve A, Isaguliants M, Falkeborn T, Sharma S, Liakina V, Robb M, Eller M, Moss B, Biberfeld G, Sandström E, Nilsson C, Markland K, Blomberg P, Wahren B. HIVIS-DNA or HIVISopt-DNA priming followed by CMDR vaccinia-based boosts induce both humoral and cellular murine immune responses to HIV. Heliyon 2017; 3:e00339. [PMID: 28721397 PMCID: PMC5496381 DOI: 10.1016/j.heliyon.2017.e00339] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 06/19/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND In order to develop a more effective prophylactic HIV-1 vaccine it is important optimize the components, improve Envelope glycoprotein immunogenicity as well as to explore prime-boost immunization schedules. It is also valuable to include several HIV-1 subtype antigens representing the world-wide epidemic. METHODS HIVIS-DNA plasmids which include Env genes of subtypes A, B and C together with Gag subtypes A and B and RTmut/Rev of subtype B were modified as follows: the Envelope sequences were shortened, codon optimized, provided with an FT4 sequence and an immunodominant region mutated. The reverse transcriptase (RT) gene was shortened to contain the most immunogenic N-terminal fragment and fused with an inactivated viral protease vPR gene. HIVISopt-DNA thus contains fewer plasmids but additional PR epitopes compared to the native HIVIS-DNA. DNA components were delivered intradermally to young Balb/c mice once, using a needle-free Biojector® immediately followed by dermal electroporation. Vaccinia-based MVA-CMDR boosts including Env gene E and Gag-RT genes A were delivered intramuscularly by needle, once or twice. RESULTS Both HIVIS-DNA and HIVISopt-DNA primed humoral and cell mediated responses well. When boosted with heterologous MVA-CMDR (subtypes A and E) virus inhibitory neutralizing antibodies were obtained to HIV-1 subtypes A, B, C and AE. Both plasmid compositions boosted with MVA-CMDR generated HIV-1 specific cellular responses directed against HIV-1 Env, Gag and Pol, as measured by IFNγ ELISpot. It was shown that DNA priming augmented the vector MVA immunological boosting effects, the HIVISopt-DNA with a trend to improved (Env) neutralization, the HIVIS-DNA with a trend to better (Gag) cell mediated immune reponses. CONCLUSIONS HIVIS-DNA was modified to obtain HIVISopt-DNA that had fewer plasmids, and additional epitopes. Even with one DNA prime followed by two MVA-CMDR boosts, humoral and cell-mediated immune responses were readily induced by priming with either DNA construct composition. Priming by HIV-DNA augmented neutralizing antibody responses revealed by boosting with the vaccinia-based heterologous sequences. Cellular and antibody responses covered selected strains representing HIV-1 subtypes A, B, C and CRF01_AE. We assume this is related to the inclusion of heterologous full genes in the vaccine schedule.
Collapse
Affiliation(s)
- J Hinkula
- Department of Clinical and Experimental Medicine, Linköping University, 58183 Linköping, Sweden
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - S Petkov
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - K Ljungberg
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - D Hallengärd
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - A Bråve
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - M Isaguliants
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - T Falkeborn
- Department of Clinical and Experimental Medicine, Linköping University, 58183 Linköping, Sweden
| | - S Sharma
- Department of Clinical and Experimental Medicine, Linköping University, 58183 Linköping, Sweden
| | - V Liakina
- Faculty of Medicine, Vilnius University 2, 08661 Vilnius, Lithuania
| | - M Robb
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, 20892 MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, 20892 MD, USA
| | - M Eller
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, 20892 MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, 20892 MD, USA
| | - B Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, 20892 MD, USA
| | - G Biberfeld
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - E Sandström
- Department of South Hospital, Karolinska Institutet, 11883 Stockholm, Sweden
| | - C Nilsson
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - K Markland
- Clinical Research Center and Vecura, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - P Blomberg
- Clinical Research Center and Vecura, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - B Wahren
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| |
Collapse
|
22
|
Shan S, Fenwick S, Ellis T, Poinern E, Edwards J, Le X, Jiang Z. Evaluation of different chemical adjuvants on an avian influenza H6 DNA vaccine in chickens. Avian Pathol 2016; 45:649-656. [PMID: 27314157 DOI: 10.1080/03079457.2016.1195488] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
This study assessed the ability of three adjuvants (aluminium hydroxide, Essai (microparticle) and Phema (nanoparticle)) to enhance the immune response of chickens to an H6N2 avian influenza DNA vaccine. No haemagglutination inhibition antibody was detected following two intramuscular immunizations with the adjuvanted and non-adjuvanted pCAG-HAk vaccine, which has previously been shown to induce moderate H6 haemagglutinin antibody response in SPF chickens. Following virus challenge, neither the vaccinated group without adjuvant nor the Essai-adjuvanted group showed a statistically significant reduction in virus shedding in oropharyngeal and cloacal swabs compared with the naive control group. However, the aluminium hydroxide and Phema-adjuvanted groups significantly reduced the frequency of virus shedding in oropharyngeal swabs, indicating that these adjuvants appeared to further enhance the vaccine potency. Aluminium hydroxide holds promise as an adjuvant for enhancing DNA-induced immune response in chickens owing to its low price and safety record.
Collapse
Affiliation(s)
- Songhua Shan
- a School of Veterinary and Life Sciences , Murdoch University , Perth , Australia
| | - Stan Fenwick
- a School of Veterinary and Life Sciences , Murdoch University , Perth , Australia
| | - Trevor Ellis
- a School of Veterinary and Life Sciences , Murdoch University , Perth , Australia
| | - Eddy Poinern
- a School of Veterinary and Life Sciences , Murdoch University , Perth , Australia
| | - John Edwards
- a School of Veterinary and Life Sciences , Murdoch University , Perth , Australia
| | - Xuan Le
- a School of Veterinary and Life Sciences , Murdoch University , Perth , Australia
| | - Zhongtao Jiang
- a School of Veterinary and Life Sciences , Murdoch University , Perth , Australia
| |
Collapse
|
23
|
Dalmia N, Klimstra WB, Mason C, Ramsay AJ. DNA-Launched Alphavirus Replicons Encoding a Fusion of Mycobacterial Antigens Acr and Ag85B Are Immunogenic and Protective in a Murine Model of TB Infection. PLoS One 2015; 10:e0136635. [PMID: 26317509 PMCID: PMC4552820 DOI: 10.1371/journal.pone.0136635] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 08/06/2015] [Indexed: 11/18/2022] Open
Abstract
There is an urgent need for effective prophylactic measures against Mycobacterium tuberculosis (Mtb) infection, particularly given the highly variable efficacy of Bacille Calmette-Guerin (BCG), the only licensed vaccine against tuberculosis (TB). Most studies indicate that cell-mediated immune responses involving both CD4+ and CD8+ T cells are necessary for effective immunity against Mtb. Genetic vaccination induces humoral and cellular immune responses, including CD4+ and CD8+ T-cell responses, against a variety of bacterial, viral, parasitic and tumor antigens, and this strategy may therefore hold promise for the development of more effective TB vaccines. Novel formulations and delivery strategies to improve the immunogenicity of DNA-based vaccines have recently been evaluated, and have shown varying degrees of success. In the present study, we evaluated DNA-launched Venezuelan equine encephalitis replicons (Vrep) encoding a novel fusion of the mycobacterial antigens α-crystallin (Acr) and antigen 85B (Ag85B), termed Vrep-Acr/Ag85B, for their immunogenicity and protective efficacy in a murine model of pulmonary TB. Vrep-Acr/Ag85B generated antigen-specific CD4+ and CD8+ T cell responses that persisted for at least 10 wk post-immunization. Interestingly, parenterally administered Vrep-Acr/Ag85B also induced T cell responses in the lung tissues, the primary site of infection, and inhibited bacterial growth in both the lungs and spleens following aerosol challenge with Mtb. DNA-launched Vrep may, therefore, represent an effective approach to the development of gene-based vaccines against TB, particularly as components of heterologous prime-boost strategies or as BCG boosters.
Collapse
MESH Headings
- Acyltransferases/genetics
- Acyltransferases/immunology
- Animals
- Antigens, Bacterial/genetics
- Antigens, Bacterial/immunology
- Bacterial Proteins/genetics
- Bacterial Proteins/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/pathology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- Disease Models, Animal
- Encephalitis Virus, Venezuelan Equine/genetics
- Encephalitis Virus, Venezuelan Equine/immunology
- Immunity, Cellular
- Immunity, Humoral
- Mice
- Mycobacterium tuberculosis/genetics
- Mycobacterium tuberculosis/immunology
- Replicon/immunology
- Tuberculosis Vaccines/genetics
- Tuberculosis Vaccines/immunology
- Tuberculosis, Pulmonary/genetics
- Tuberculosis, Pulmonary/immunology
- Tuberculosis, Pulmonary/pathology
- Tuberculosis, Pulmonary/prevention & control
- Vaccination
- alpha-Crystallins/genetics
- alpha-Crystallins/immunology
Collapse
Affiliation(s)
- Neha Dalmia
- Department of Microbiology, Immunology and Parasitology, LSU Health Sciences Center, New Orleans, Louisiana, United States of America
| | - William B. Klimstra
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Carol Mason
- Department of Microbiology, Immunology and Parasitology, LSU Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Alistair J. Ramsay
- Department of Microbiology, Immunology and Parasitology, LSU Health Sciences Center, New Orleans, Louisiana, United States of America
- Department of Microbiology, Immunology and Parasitology, LSU Health Sciences Center, New Orleans, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
24
|
Xie X, Wang L, Yang W, Yu R, Li Q, Pang X. Co-administration of antigen with chemokine MCP-3 or MDC/CCL22 enhances DNA vaccine potency. Invest New Drugs 2015; 33:810-5. [DOI: 10.1007/s10637-015-0250-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 05/01/2015] [Indexed: 10/23/2022]
|
25
|
Wang HC, An HJ, Yu YZ, Xu Q. Potentiation of anthrax vaccines using protective antigen-expressing viral replicon vectors. Immunol Lett 2015; 163:206-13. [DOI: 10.1016/j.imlet.2014.07.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 07/18/2014] [Accepted: 07/27/2014] [Indexed: 11/17/2022]
|
26
|
Kamhawi S, Oliveira F, Valenzuela JG. Using humans to make a human leishmaniasis vaccine. Sci Transl Med 2014; 6:234fs18. [PMID: 24786322 DOI: 10.1126/scitranslmed.3009118] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The cellular immune response to peptide pools from conserved Leishmania antigens in leishmaniasis-immune individuals identified epitopes for a human DNA vaccine (Das et al., this issue).
Collapse
Affiliation(s)
- Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | | | | |
Collapse
|
27
|
Yu YZ, Ma Y, Xu WH, Wang S, Sun ZW. Combinations of various CpG motifs cloned into plasmid backbone modulate and enhance protective immunity of viral replicon DNA anthrax vaccines. Med Microbiol Immunol 2014; 204:481-91. [PMID: 25265876 DOI: 10.1007/s00430-014-0359-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 09/16/2014] [Indexed: 10/24/2022]
Abstract
DNA vaccines are generally weak stimulators of the immune system. Fortunately, their efficacy can be improved using a viral replicon vector or by the addition of immunostimulatory CpG motifs, although the design of these engineered DNA vectors requires optimization. Our results clearly suggest that multiple copies of three types of CpG motifs or combinations of various types of CpG motifs cloned into a viral replicon vector backbone with strong immunostimulatory activities on human PBMC are efficient adjuvants for these DNA vaccines to modulate and enhance protective immunity against anthrax, although modifications with these different CpG forms in vivo elicited inconsistent immune response profiles. Modification with more copies of CpG motifs elicited more potent adjuvant effects leading to the generation of enhanced immunity, which indicated a CpG motif dose-dependent enhancement of antigen-specific immune responses. Notably, the enhanced and/or synchronous adjuvant effects were observed in modification with combinations of two different types of CpG motifs, which provides not only a contribution to the knowledge base on the adjuvant activities of CpG motifs combinations but also implications for the rational design of optimal DNA vaccines with combinations of CpG motifs as "built-in" adjuvants. We describe an efficient strategy to design and optimize DNA vaccines by the addition of combined immunostimulatory CpG motifs in a viral replicon DNA plasmid to produce strong immune responses, which indicates that the CpG-modified viral replicon DNA plasmid may be desirable for use as vector of DNA vaccines.
Collapse
Affiliation(s)
- Yun-Zhou Yu
- Beijing Institute of Biotechnology, 20 Dongdajie Street, Fengtai District, Beijing, 100071, China,
| | | | | | | | | |
Collapse
|
28
|
Jones CH, Hakansson AP, Pfeifer BA. Biomaterials at the interface of nano- and micro-scale vector-cellular interactions in genetic vaccine design. J Mater Chem B 2014; 46:8053-8068. [PMID: 29887986 PMCID: PMC5990286 DOI: 10.1039/c4tb01058b] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The development of safe and effective vaccines for the prevention of elusive infectious diseases remains a public health priority. Immunization, characterized by adaptive immune responses to specific antigens, can be raised by an array of delivery vectors. However, current commercial vaccination strategies are predicated on the retooling of archaic technology. This review will discuss current and emerging strategies designed to elicit immune responses in the context of genetic vaccination. Selected strategies at the biomaterial-biological interface will be emphasized to illustrate the potential of coupling both fields towards a common goal.
Collapse
Affiliation(s)
- Charles H Jones
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260-4200, USA
| | - Anders P Hakansson
- Department of Microbiology and Immunology, University at Buffalo, The State University of New York, Buffalo, NY 14260-4200, USA
- The Witebsky Center for Microbial Pathogenesis and Immunology, University at Buffalo, The State University of New York, Buffalo, NY 14260-4200, USA
| | - Blaine A Pfeifer
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260-4200, USA
| |
Collapse
|
29
|
Duthie MS, Reed SG. The Emergence of Defined Subunit Vaccines for the Prevention of Leishmaniasis. CURRENT TROPICAL MEDICINE REPORTS 2014. [DOI: 10.1007/s40475-014-0024-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
30
|
Ullas PT, Desai A, Madhusudana SN. Immunogenicity and efficacy of a plasmid DNA rabies vaccine incorporating Myd88 as a genetic adjuvant. Clin Exp Vaccine Res 2014; 3:202-11. [PMID: 25003094 PMCID: PMC4083073 DOI: 10.7774/cevr.2014.3.2.202] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 04/29/2014] [Accepted: 05/01/2014] [Indexed: 11/15/2022] Open
Abstract
PURPOSE Myeloid differentiation factor 88 (Myd88), a ubiquitous Toll-like receptor adaptor molecule, has been reported to play important roles in B cell responses to infections and vaccination. The present study evaluated the effects of genetic adjuvanting with Myd88 on the immune responses to a plasmid DNA rabies vaccine. MATERIALS AND METHODS Plasmids encoding rabies glycoprotein alone (pIRES-Rgp) or a fragment of Myd88 gene in addition (pIRES-Rgp-Myd) were constructed and administered intramuscularly or intrademally in Swiss albino mice (on days 0, 7, and 21). Rabies virus neutralizing antibody (RVNA) titres were estimated in the mice sera on days 14 and 28 by rapid fluorescent focus inhibition test. The protective efficacy of the constructs was evaluated by an intracerebral challenge with challenge virus standard virus on day 35. RESULTS Co-expression of Myd88 increased RVNA responses to pIRES-Rgp by 3- and 2-folds, following intramuscular and intradermal immunization, respectively. pIRES-Rgp protected 80% of the mice following intramuscular and intradermal immunizations, while pIRES-Rgp-Myd afforded 100% protection following similar administrations. CONCLUSION Genetic adjuvanting with Myd88 enhanced the RVNA responses and protective efficacy of a plasmid DNA rabies vaccine. This strategy might be useful for rabies vaccination of canines in the field, and needs further evaluation.
Collapse
Affiliation(s)
| | - Anita Desai
- Department of Neurovirology, National Institute of Mental Health and Neurosciences, Bangalore, Karnataka, India
| | - Shampur Narayan Madhusudana
- Department of Neurovirology, National Institute of Mental Health and Neurosciences, Bangalore, Karnataka, India
| |
Collapse
|
31
|
van Drunen Littel-van den Hurk S, Hannaman D. Electroporation for DNA immunization: clinical application. Expert Rev Vaccines 2014; 9:503-17. [DOI: 10.1586/erv.10.42] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
32
|
Saljoughian N, Zahedifard F, Doroud D, Doustdari F, Vasei M, Papadopoulou B, Rafati S. Cationic solid-lipid nanoparticles are as efficient as electroporation in DNA vaccination against visceral leishmaniasis in mice. Parasite Immunol 2013; 35:397-408. [DOI: 10.1111/pim.12042] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 05/23/2013] [Indexed: 02/03/2023]
Affiliation(s)
- N. Saljoughian
- Molecular Immunology and Vaccine Research Laboratory; Pasteur Institute of Iran; Tehran Iran
| | - F. Zahedifard
- Molecular Immunology and Vaccine Research Laboratory; Pasteur Institute of Iran; Tehran Iran
| | - D. Doroud
- Department of Quality Control; Research and Production Complex; Pasteur Institute of Iran; Tehran Iran
| | - F. Doustdari
- Molecular Immunology and Vaccine Research Laboratory; Pasteur Institute of Iran; Tehran Iran
| | - M. Vasei
- Department of Pathology; Shariati Hospital; Tehran University of Medical Sciences; Tehran Iran
| | - B. Papadopoulou
- Research Centre in Infectious Disease; CHU de Quebec Research Centre (CHUL); Quebec QC Canada
- Department of Microbiology; Infectious Disease and Immunology; Faculty of Medicine; Laval University; Quebec QC Canada
| | - S. Rafati
- Molecular Immunology and Vaccine Research Laboratory; Pasteur Institute of Iran; Tehran Iran
| |
Collapse
|
33
|
Shima F, Uto T, Akagi T, Baba M, Akashi M. Size effect of amphiphilic poly(γ-glutamic acid) nanoparticles on cellular uptake and maturation of dendritic cells in vivo. Acta Biomater 2013; 9:8894-901. [PMID: 23770225 DOI: 10.1016/j.actbio.2013.06.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Revised: 05/30/2013] [Accepted: 06/07/2013] [Indexed: 01/13/2023]
Abstract
We prepared size-regulated nanoparticles (NPs) composed of amphiphilic poly(γ-glutamic acid) (γ-PGA). In this study, 40, 100 and 200 nm γ-PGA-graft-l-phenylalanine ethylester (γ-PGA-Phe) NPs were employed. The size of NPs significantly influenced the uptake and activation behaviors of antigen-presenting cells (APCs). When 40 nm γ-PGA-Phe NPs were applied to these cells in vitro, they were highly activated compared with 100 and 200 nm NPs, while cellular uptake was size dependent. The size of the γ-PGA-Phe NPs also significantly affected their migration to the lymph nodes and uptake behavior of NPs by dendritic cells (DCs) in vivo. The 40 nm γ-PGA-Phe NPs migrated more rapidly to the lymph nodes and were taken up by a greater number of DCs compared with 100 and 200 nm NPs. On the other hand, when the amount of γ-PGA-Phe NPs taken up per DC was evaluated, it was higher for 100 and 200 nm NPs than for 40 nm NPs, which suggests that the larger γ-PGA-Phe NPs can deliver a large amount of antigen to a single DC compared with smaller NPs. Furthermore, when examined the maturation of DCs in lymph nodes, 40 nm γ-PGA-Phe NPs efficiently stimulated DCs. These results suggest that the activation, uptake behavior by APCs, migration to lymph nodes, and DC maturation can be controlled by the size of γ-PGA-Phe NPs.
Collapse
Affiliation(s)
- Fumiaki Shima
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Japan; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Kawaguchi Center Building, 4-1-8 Honcho, Kawaguchi 332-0012, Japan
| | | | | | | | | |
Collapse
|
34
|
Hong X, Wei L, Wu F, Wu Z, Chen L, Liu Z, Yuan W. Dissolving and biodegradable microneedle technologies for transdermal sustained delivery of drug and vaccine. DRUG DESIGN DEVELOPMENT AND THERAPY 2013; 7:945-52. [PMID: 24039404 PMCID: PMC3771849 DOI: 10.2147/dddt.s44401] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Microneedles were first conceptualized for drug delivery many decades ago, overcoming the shortages and preserving the advantages of hypodermic needle and conventional transdermal drug-delivery systems to some extent. Dissolving and biodegradable microneedle technologies have been used for transdermal sustained deliveries of different drugs and vaccines. This review describes microneedle geometry and the representative dissolving and biodegradable microneedle delivery methods via the skin, followed by the fabricating methods. Finally, this review puts forward some perspectives that require further investigation.
Collapse
Affiliation(s)
- Xiaoyun Hong
- Department of Neurology, Xinhua Hospital, Shanghai JiaoTong University School of Medicine, 1665 Kongjiang Road, Shanghai, People’s Republic of China
| | | | | | | | | | | | | |
Collapse
|
35
|
DNA vaccines targeting the encoded antigens to dendritic cells induce potent antitumor immunity in mice. BMC Immunol 2013; 14:39. [PMID: 23941509 PMCID: PMC3751307 DOI: 10.1186/1471-2172-14-39] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 07/31/2013] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Although DNA vaccine holds a great potential for cancer immunotherapy, effective long-lasting antitumoral immunity sufficient to induce durable responses in cancer patients remains to be achieved. Considering the pivotal role of dendritic cells (DC) in the antigen processing and presentation, we prepared DC-targeting DNA vaccines by fusing tumor-associated antigen HER2/neu ectodomain to single chain antibody fragment (scFv) from NLDC-145 antibody specific for DC-restricted surface molecule DEC-205 (scFvNLDC-145), and explored its antitumoral efficacy and underlying mechanisms in mouse breast cancer models. RESULTS In vivo targeting assay demonstrated that scFvNLDC-145 specifically delivered DNA vaccine-encoded antigen to DC. Compared with untargeted HER2/neu DNA vaccines, vaccination with scFvNLDC-145-HER2/neu markedly promoted the HER2/neu-specific cellular and humoral immune responses with long-lasting immune memory, resulting in effective protection against challenge of HER2/neu-positive D2F2/E2 breast tumor while ineffective in parental HER2/neu-negative D2F2 breast tumor. More importantly, in combination with temporary depletion of regulatory T cells (Treg) by low-dose cyclophosphamide, vaccination with scFvNLDC-145-HER2/neu induced the regression of established D2F2/E2 breast tumor and significantly retarded the development of spontaneous mammary carcinomas in transgenic BALB-neuT mice. CONCLUSION Our findings demonstrate that DC-targeted DNA vaccines for in vivo direct delivery of tumor antigens to DC could induce potent antigen-specific cellular and humoral immune responses and, if additional combination with systemic Treg depletion, was able to elicit an impressively therapeutic antitumoral activity, providing a rationale for further development of this approach for cancer treatment.
Collapse
|
36
|
Li P, Chen S, Jiang Y, Jiang J, Zhang Z, Sun X. Dendritic cell targeted liposomes-protamine-DNA complexes mediated by synthetic mannosylated cholesterol as a potential carrier for DNA vaccine. NANOTECHNOLOGY 2013; 24:295101. [PMID: 23799649 DOI: 10.1088/0957-4484/24/29/295101] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
To construct mannosylated liposomes/protamine/DNA (LPD) carriers for DNA vaccine targeting to dendritic cells (DCs), a mannosylated cholesterol derivative (Man-C6-Chol) was synthesized via simple ester linkage and amide bonds. Then, the Man-C6-Chol was applied to LPD formulation as a synthetic ligand. The physicochemical properties of mannosylated LPD (Man-LPD) were first evaluated, including the size and zeta potential, morphology and the ability to protect DNA against DNase I degradation. Man-LPD showed a small size with a stable viral-like structure. In comparison to non-mannose liposomes/LPD (Man-free liposomes/LPD), mannosylated liposomes/LPD (Man-liposomes/Man-LPD) exhibited higher efficiency in both intracellular uptake (2.3-fold) and transfection (4.5-fold) in vitro. Subsequent MTT assays indicated that the LPD carriers had low toxicity on the tested cells. Afterwards, the investigation into the maturation activation on primary bone marrow-derived DCs (BMDCs) showed that both Man-LPD and Man-free LPD induced remarkable up-regulation of CD80, CD86 and CD40 on BMDCs. Inspired by these studies, we can conclude that the synthetic mannosylated LPD targeting to DCs was a potential carrier for DNA vaccine.
Collapse
Affiliation(s)
- Pan Li
- Key Laboratory of Drug Targeting and Novel Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | | | | | | | | | | |
Collapse
|
37
|
Abstract
Biotechnology uses substances, materials or extracts derived from living cells, employing 22 million Europeans in a € 1.5 Tn endeavour, being the premier global economic growth opportunity this century. Significant advances have been made in red biotechnology using pharmaceutically and medically relevant applications, green biotechnology developing agricultural and environmental tools and white biotechnology serving industrial scale uses, frequently as process feedstocks. Red biotechnology has delivered dramatic improvements in controlling human disease, from antibiotics to overcome bacterial infections to anti-HIV/AIDS pharmaceuticals such as azidothymidine (AZT), anti-malarial compounds and novel vaccines saving millions of lives. Green biotechnology has dramatically increased food production through Agrobacterium and biolistic genetic modifications for the development of 'Golden Rice', pathogen resistant crops expressing crystal toxin genes, drought resistance and cold tolerance to extend growth range. The burgeoning area of white biotechnology has delivered bio-plastics, low temperature enzyme detergents and a host of feedstock materials for industrial processes such as modified starches, without which our everyday lives would be much more complex. Biotechnological applications can bridge these categories, by modifying energy crops properties, or analysing circulating nucleic acid elements, bringing benefits for all, through increased food production, supporting climate change adaptation and the low carbon economy, or novel diagnostics impacting on personalized medicine and genetic disease. Cross-cutting technologies such as PCR, novel sequencing tools, bioinformatics, transcriptomics and epigenetics are in the vanguard of biotechnological progress leading to an ever-increasing breadth of applications. Biotechnology will deliver solutions to unimagined problems, providing food security, health and well-being to mankind for centuries to come.
Collapse
|
38
|
Diehl MC, Lee JC, Daniels SE, Tebas P, Khan AS, Giffear M, Sardesai NY, Bagarazzi ML. Tolerability of intramuscular and intradermal delivery by CELLECTRA(®) adaptive constant current electroporation device in healthy volunteers. Hum Vaccin Immunother 2013; 9:2246-52. [PMID: 24051434 DOI: 10.4161/hv.24702] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
DNA vaccines are being developed as a potentially safe and effective immunization platform. However, translation of DNA vaccines into a clinical setting has produced results that have fallen short of those generated in a preclinical setting. Various strategies are being developed to address this lack of potency, including improvements in delivery methods. Electroporation (EP) creates transient increases in cell membrane permeability, thus enhancing DNA uptake and leading to a more robust immune response. Here, we report on the safety and tolerability of delivering sterile saline via intramuscular (IM) or intradermal (ID) injection followed by in vivo electroporation using the CELLECTRA(®) adaptive constant current device in healthy adults from two open-label studies. Pain, as assessed by VAS, was highest immediately after EP but diminishes by about 50% within 5 min. Mean VAS scores appear to correlate with the amount of energy delivered and depth of needle insertion, especially for intramuscular EP. Mean scores did not exceed 7 out of 10 or 3 out of 10 for IM and ID EP, respectively. The majority of adverse events included mild to moderate injection site reactions that resolved within one day. No deaths or serious adverse events were reported during the course of either study. Overall, injection followed by EP with the CELLECTRA(®) device was well-tolerated and no significant safety concerns were identified. These studies support the further development of electroporation as a vaccine delivery method to enhance immunogenicity, particularly for diseases in which traditional vaccination approaches are ineffective.
Collapse
Affiliation(s)
| | | | | | - Pablo Tebas
- University of Pennsylvania; Division of Infectious Disease; Philadelphia, PA USA
| | | | | | | | | |
Collapse
|
39
|
DeMuth PC, Min Y, Huang B, Kramer JA, Miller AD, Barouch DH, Hammond PT, Irvine DJ. Polymer multilayer tattooing for enhanced DNA vaccination. NATURE MATERIALS 2013; 12:367-76. [PMID: 23353628 PMCID: PMC3965298 DOI: 10.1038/nmat3550] [Citation(s) in RCA: 213] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 12/13/2012] [Indexed: 05/19/2023]
Abstract
DNA vaccines have many potential benefits but have failed to generate robust immune responses in humans. Recently, methods such as in vivo electroporation have demonstrated improved performance, but an optimal strategy for safe, reproducible, and pain-free DNA vaccination remains elusive. Here we report an approach for rapid implantation of vaccine-loaded polymer films carrying DNA, immune-stimulatory RNA, and biodegradable polycations into the immune-cell-rich epidermis, using microneedles coated with releasable polyelectrolyte multilayers. Films transferred into the skin following brief microneedle application promoted local transfection and controlled the persistence of DNA and adjuvants in the skin from days to weeks, with kinetics determined by the film composition. These 'multilayer tattoo' DNA vaccines induced immune responses against a model HIV antigen comparable to electroporation in mice, enhanced memory T-cell generation, and elicited 140-fold higher gene expression in non-human primate skin than intradermal DNA injection, indicating the potential of this strategy for enhancing DNA vaccination.
Collapse
Affiliation(s)
- Peter C DeMuth
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts 02139, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Sun K, Li X, Jiang J, Cheng A, Wang M, Zhu D, Jia R, Chen S, Zhou Y, Chen X, Wang X. Distribution characteristics of DNA vaccine encoded with glycoprotein C from Anatid herpesvirus 1 with chitosan and liposome as deliver carrier in ducks. Virol J 2013; 10:89. [PMID: 23497107 PMCID: PMC3616852 DOI: 10.1186/1743-422x-10-89] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2012] [Accepted: 03/07/2013] [Indexed: 12/03/2022] Open
Abstract
Background A eukaryotic expression plasmid encoding glycoprotein C (gC) of Anatid herpesvirus 1 (AnHV-1) (pcDNA3.1-gC) was constructed and validated. The tissue distribution of chitosan/DNA complexes, liposome/DNA complexes and pcDNA3.1-gC alone were evaluated using a quantitative real-time PCR based TaqMan™ probe following intramuscular administration in ducklings. Results Compared with pcDNA3.1-gC alone, liposomes universally increased the plasmid DNA copy number at the injection sites, liver, spleen, heart, brain, bursa of Fabricius, and especially in the enteron (esophagus, duodenum, rectum, and cecum). Chitosan also universally increased the plasmid DNA copy number at the injection sites, liver, spleen, heart, brain and esophagus. Compared with lipoplex-gC, higher chitosan-gC plasmid DNA copy numbers were detected at the injection sites, liver, spleen, heart, brain and esophagus. In contrast, compared with lipoplex-gC, lower copy numbers of chitosan-gC plasmid DNA were detected in the duodenum, rectum and cecum. Conclusions The results of this study demonstrated that chitosan and liposomes mediated rapid and extensive plasmid distribution in duck tissues, with low levels maintained from 1 d after DNA vaccination.
Collapse
Affiliation(s)
- Kunfeng Sun
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan, 611130, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Ma Y, An HJ, Wei XQ, Xu Q, Yu YZ, Sun ZW. Enhanced potency of replicon vaccine using one vector to simultaneously co-express antigen and interleukin-4 molecular adjuvant. Hum Vaccin Immunother 2013; 9:242-9. [PMID: 23291932 DOI: 10.4161/hv.22888] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
We evaluated the utility of interleukin-4 (IL-4) as molecular adjuvant of replicon vaccines for botulinum neurotoxin serotype A (BoNT/A) in mouse model. In both Balb/c and C57/BL6 mice that received the plasmid DNA replicon vaccines derived from Semliki Forest virus (SFV) encoding the Hc gene of BoNT/A (AHc), the immunogenicity was significantly modulated and enhanced by co-delivery or co-express of the IL-4 molecular adjuvant. The enhanced potencies were also produced by co-delivery or co-expression of the IL-4 molecular adjuvant in mice immunized with the recombinant SFV replicon particles (VRP) vaccines. In particular, when AHc and IL-4 were co-expressed within the same replicon vaccine vector using dual-expression or bicistronic IRES, the anti-AHc antibody titers, serum neutralization titers and survival rates of immunized mice after challenged with BoNT/A were significantly increased. These results indicate IL-4 is an effective Th2-type adjuvant for the replicon vaccines in both strain mice, and the co-expression replicon vaccines described here may be an excellent candidate for further vaccine development in other animals or humans. Thus, we described a strategy to design and develop efficient vaccines against BoNT/A or other pathogens using one replicon vector to simultaneously co-express antigen and molecular adjuvant.
Collapse
Affiliation(s)
- Yao Ma
- College of Life Science and Bioengineering; Beijing Jiaotong University; Beijing, PR China
| | | | | | | | | | | |
Collapse
|
42
|
Immunomodulatory effects of IP-10 chemokine along with PEI600-Tat delivery system in DNA vaccination against HPV infections. Mol Immunol 2013; 53:149-60. [DOI: 10.1016/j.molimm.2012.07.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2012] [Revised: 07/18/2012] [Accepted: 07/26/2012] [Indexed: 12/23/2022]
|
43
|
The development of gene-based vectors for immunization. Vaccines (Basel) 2013. [PMCID: PMC7151937 DOI: 10.1016/b978-1-4557-0090-5.00064-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
44
|
Todolí F, Rodríguez-Cortés A, Núñez MDC, Laurenti MD, Gómez-Sebastián S, Rodríguez F, Pérez-Martín E, Escribano JM, Alberola J. Head-to-head comparison of three vaccination strategies based on DNA and raw insect-derived recombinant proteins against Leishmania. PLoS One 2012; 7:e51181. [PMID: 23236448 PMCID: PMC3517401 DOI: 10.1371/journal.pone.0051181] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Accepted: 10/30/2012] [Indexed: 12/14/2022] Open
Abstract
Parasitic diseases plague billions of people among the poorest, killing millions annually, and causing additional millions of disability-adjusted life years lost. Leishmaniases affect more than 12 million people, with over 350 million people at risk. There is an urgent need for efficacious and cheap vaccines and treatments against visceral leishmaniasis (VL), its most severe form. Several vaccination strategies have been proposed but to date no head-to-head comparison was undertaken to assess which is the best in a clinical model of the disease. We simultaneously assayed three vaccination strategies against VL in the hamster model, using KMPII, TRYP, LACK, and PAPLE22 vaccine candidate antigens. Four groups of hamsters were immunized using the following approaches: 1) raw extracts of baculovirus-infected Trichoplusia ni larvae expressing individually one of the four recombinant proteins (PROT); 2) naked pVAX1 plasmids carrying the four genes individually (DNA); 3) a heterologous prime-boost (HPB) strategy involving DNA followed by PROT (DNA-PROT); and 4) a Control including empty pVAX1 plasmid followed by raw extract of wild-type baculovirus-infected T. ni larvae. Hamsters were challenged with L. infantum promastigotes and maintained for 20 weeks. While PROT vaccine was not protective, DNA vaccination achieved protection in spleen. Only DNA-PROT vaccination induced significant NO production by macrophages, accompanied by a significant parasitological protection in spleen and blood. Thus, the DNA-PROT strategy elicits strong immune responses and high parasitological protection in the clinical model of VL, better than its corresponding naked DNA or protein versions. Furthermore, we show that naked DNA coupled with raw recombinant proteins produced in insect larvae biofactories -the cheapest way of producing DNA-PROT vaccines- is a practical and cost-effective way for potential "off the shelf" supplying vaccines at very low prices for the protection against leishmaniases, and possibly against other parasitic diseases affecting the poorest of the poor.
Collapse
Affiliation(s)
- Felicitat Todolí
- LeishLAB–Servei d’Anàlisi de Fàrmacs, Departament de Farmacologia, de Terapèutica i de Toxicologia, Edifici V, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Alhelí Rodríguez-Cortés
- LeishLAB–Servei d’Anàlisi de Fàrmacs, Departament de Farmacologia, de Terapèutica i de Toxicologia, Edifici V, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - María del Carmen Núñez
- Alternative Gene Expression S.L., Centro Empresarial, Parque Científico y Tecnológico de la Universidad Politécnica de Madrid, Pozuelo de Alarcón, Madrid, Spain
| | - Márcia D. Laurenti
- Laboratorio Patologia de Moléstias Infecciosas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Silvia Gómez-Sebastián
- Alternative Gene Expression S.L., Centro Empresarial, Parque Científico y Tecnológico de la Universidad Politécnica de Madrid, Pozuelo de Alarcón, Madrid, Spain
| | - Fernando Rodríguez
- Centre de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Campus de la UAB, Bellaterra, Barcelona, Spain
| | - Eva Pérez-Martín
- Centre de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Campus de la UAB, Bellaterra, Barcelona, Spain
| | - José M. Escribano
- Departamento de Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Madrid, Spain
| | - Jordi Alberola
- LeishLAB–Servei d’Anàlisi de Fàrmacs, Departament de Farmacologia, de Terapèutica i de Toxicologia, Edifici V, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| |
Collapse
|
45
|
Yu YZ, Li N, Ma Y, Wang S, Yu WY, Sun ZW. Three types of human CpG motifs differentially modulate and augment immunogenicity of nonviral and viral replicon DNA vaccines as built-in adjuvants. Eur J Immunol 2012; 43:228-39. [PMID: 23037552 DOI: 10.1002/eji.201242690] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 09/14/2012] [Accepted: 10/01/2012] [Indexed: 01/20/2023]
Abstract
NakedDNA vaccines given by intramuscular injection are efficient in mouse models, but they require improvement for human use. As the immunogenicity of DNA vaccines depends, to a large extent, on the presence of CpG motifs as built-in adjuvants, we addressed this issue by inserting three types of human CpG motifs (A-type, B-type, and C-type) into the backbone of nonviral DNA and viral DNA replicon vectors with distinct immunostimulatory activities on human PBMCs. The adjuvant effects of CpG modifications in DNA vaccines expressing three types of antigens (β-Gal, AHc, or PA4) were then characterized in mice and found to significantly enhance antigen-specific humoral and cell-mediated immune responses. The three types of CpG motifs also differentially affected and modulated immune responses and protective potency against botulinum neurotoxin serotype A and Bacillus anthracis A16R challenge. Taken together, these results demonstrate that insertion of human CpG motifs can differentially modulate the immunogenicity of nonviral DNA vaccines as well as viral DNA replicon vaccines. Our study provides not only a better understanding of the in vivo activities of CpG motif adjuvants but implications for the rational design of such motifs as built-in adjuvants for DNA vectors targeting specific antigens.
Collapse
Affiliation(s)
- Yun-Zhou Yu
- Beijing Institute of Biotechnology, Beijing, China.
| | | | | | | | | | | |
Collapse
|
46
|
Spång HCL, Braathen R, Bogen B. Heterodimeric barnase-barstar vaccine molecules: influence of one versus two targeting units specific for antigen presenting cells. PLoS One 2012; 7:e45393. [PMID: 23028981 PMCID: PMC3445521 DOI: 10.1371/journal.pone.0045393] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 08/22/2012] [Indexed: 11/30/2022] Open
Abstract
It is known that targeting of antigen to antigen presenting cells (APC) increases immune responses. However, it is unclear if more than one APC-specific targeting unit in the antigenic molecule will increase responses. To address this issue, we have here made heterodimeric vaccine molecules that each express four different fusion subunits. The bacterial ribonuclease barnase and its inhibitor barstar interact with high affinity, and the barnase-barstar complex was therefore used as a dimerization unit. Barnase and barstar were fused N-terminally with single chain fragment variable (scFv)s targeting units specific for either MHC class II molecules on APC or the hapten 5-iodo-4-hydroxy-3-nitrophenylacetyl (NIP). C-terminal antigenic fusions were either the fluorescent protein mCherry or scFv315 derived from myeloma protein M315. The heterodimeric vaccine molecules were formed both in vitro and in vivo. Moreover, the four different fused moieties appeared to fold correctly since they retained their specificity and function. DNA vaccination with MHC class II-targeted vaccine induced higher mCherry-specific IgG1 responses compared to non-targeted control. Since mCherry and MHC class II are in trans in this heterodimer, this suggests that heterodimeric proteins are formed in vivo without prior protein purification. Surprisingly, one targeting moiety was sufficient for the increased IgG1 response, and addition of a second targeting moiety did not increase responses. Similar results were found in in vitro T cell assays; vaccine molecules with one targeting unit were as potent as those with two. In combination with the easy cloning strategy, the heterodimeric barnase-barstar vaccine molecule could provide a flexible platform for development of novel DNA vaccines with increased potency.
Collapse
Affiliation(s)
- Heidi Cecilie Larsen Spång
- Centre for Immune Regulation, Institute of Immunology, University of Oslo and Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Ranveig Braathen
- Centre for Immune Regulation, Institute of Immunology, University of Oslo and Oslo University Hospital, Rikshospitalet, Oslo, Norway
- * E-mail:
| | - Bjarne Bogen
- Centre for Immune Regulation, Institute of Immunology, University of Oslo and Oslo University Hospital, Rikshospitalet, Oslo, Norway
| |
Collapse
|
47
|
Song JM, Lee KH, Seong BL. Recombinant influenza viruses as delivery vectors for hepatis B virus epitopes. Clin Exp Vaccine Res 2012; 1:77-82. [PMID: 23596580 PMCID: PMC3623514 DOI: 10.7774/cevr.2012.1.1.77] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2012] [Revised: 06/08/2012] [Accepted: 06/15/2012] [Indexed: 12/31/2022] Open
Abstract
Purpose Neuraminidase (NA) of influenza virus contains stalk region that shows a great deal of variability in both amino acid sequence and length. In this paper, we investigated generation of recombinant influenza viruses that had hepatitis B virus (HBV) B cell epitopes in the NA stalk region as a dual vaccine candidate. Materials and Methods We used the WSH-HK reassortant helper virus for rescue of recombinant influenza virus containing HBV epitopes and reverse genetic protocol based on the use of micrococcal nuclease-treated virus cores for reconstitution of ribonucleoproteins. Results We successfully generated a chimeric influenza viruses which contained 22 amino acid peptides in the stalk region derived from the surface and pre-surface protein HBV. The growth kinetics of the recombinant viruses was investigated after infection of Madin-Darby canine kidney (MDCK) and Madin-Darby bovine kidney (MDBK) cells and the rIV-BVPreS virus showed higher titer than other viruses in MDCK cells. We also confirmed the presence of HBV epitopes in the chimeric viruses by enzyme-linked immunosorbent assay (ELISA) using anti-HBV polyclonal antibody. When the ratio of recombinant virus verse wild type virus was calculated by ELISA, recombinant viruses exhibited 2 fold higher values than the wild type virus. Conclusion These results suggest that chimeric influenza virus which contained foreign antigens can be used as dual vaccine against both HBV and influenza viruses.
Collapse
Affiliation(s)
- Jae-Min Song
- Department of Global Medical Science, College of Nursing, Sungshin Women's University, Seoul, Korea
| | | | | |
Collapse
|
48
|
Ullah S, Shah MAA, Riaz N. Recent Advances in Development of DNA Vaccines Against Hepatitis C virus. INDIAN JOURNAL OF VIROLOGY : AN OFFICIAL ORGAN OF INDIAN VIROLOGICAL SOCIETY 2012; 23:253-60. [PMID: 24293811 DOI: 10.1007/s13337-012-0058-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Accepted: 02/03/2012] [Indexed: 01/28/2023]
Abstract
Hepatitis C is one of the foremost challenging diseases all over the world. No vaccine has been developed, yet against Hepatitis C virus (HCV). This is partly due to the high mutation rate in the HCV genome, which generates new genotypes and sub genotypes. A mass of efforts have been devoted for the development of an efficient vaccine against HCV. DNA Vaccines, an emerging field of Vaccinology, grasp strong potential to be the most reliable and efficient mode of vaccination in the future. This technology is under investigation currently. Incredibly diverse approaches have been applied as an endeavor to develop a potent DNA vaccine against HCV. The HCV structural genes and the virus like particles have been attempted and so far the results are quite promising in the Lab animals. As there is no proper animal model for HCV infection except chimpanzees, it is very difficult to articulate whether these vaccines will also be pertinent in humans or not. This review will focus on different approaches being used for the development of DNA vaccines, the major tribulations in designing a DNA vaccine against HCV as well as the future prospects for the improvement of under trials DNA vaccines developed against HCV.
Collapse
Affiliation(s)
- Sami Ullah
- NUST Center of Virology and Immunology, National University of Science and Technology, Islamabad, Pakistan
| | | | | |
Collapse
|
49
|
Hanif SNM, Al-Attiyah R, Mustafa AS. Cellular immune responses in mice induced by M. tuberculosis PE35-DNA vaccine construct. Scand J Immunol 2011; 74:554-60. [PMID: 21812801 DOI: 10.1111/j.1365-3083.2011.02604.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The PE35 (Rv3872) gene of Mycobacterium tuberculosis is present in the region of difference (RD) one that is deleted in all vaccine strains of Mycobacterium bovis bacillus Calmette Guerin. The aim of this study was to clone PE35 DNA into a DNA vaccine plasmid with CMV promoter and interleukin-2 secretory signal and evaluate the recombinant plasmid for induction of antigen-specific cellular responses in mice. DNA corresponding to PE35 was PCR amplified from the genomic DNA of M. tuberculosis H(37) Rv, cloned into pGEMT-Easy vector and sub-cloned into the DNA vaccine vector pUMVC6. BALB/c mice were immunized with recombinant pUMVC6/PE35 and spleen cells were tested for T-helper (Th)1-type (antigen-induced proliferation and secretion of IFN-γ) and Th2-type (IL-5), and anti-inflammatory (IL-10) cytokine responses to pure recombinant PE35 protein and its synthetic peptides. Mice immunized with the recombinant plasmid DNA (pUMVC6/PE35) showed positive Th1-type cellular responses to pure PE35, but not to an irrelevant antigen, i.e. PPE68 (Rv3873). However, the vaccine construct did not induce antigen-specific Th2-type (IL-5) or anti-inflammatory (IL-10) reactivity to PE35. Testing with synthetic peptides showed that Th1-type cells recognizing various epitopes of PE35 were induced in mice immunized with pUMVC6/PE35 DNA. These results suggest that pUMVC6/PE35 may be useful as a safer vaccine candidate against TB.
Collapse
Affiliation(s)
- S N M Hanif
- Faculty of Medicine, Department of Microbiology, Faculty of Medicine, Kuwait University, Safat, Kuwait.
| | | | | |
Collapse
|
50
|
Carvalho GBF, Silva-Pereira RAD, Pacífico LGG, Fonseca CT. Identification of Schistosoma mansoni candidate antigens for diagnosis of schistosomiasis. Mem Inst Oswaldo Cruz 2011; 106:837-43. [DOI: 10.1590/s0074-02762011000700009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Accepted: 07/26/2011] [Indexed: 01/22/2023] Open
|