1
|
Marquez-Martinez S, Salisch N, Serroyen J, Zahn R, Khan S. Peak transgene expression after intramuscular immunization of mice with adenovirus 26-based vector vaccines correlates with transgene-specific adaptive immune responses. PLoS One 2024; 19:e0299215. [PMID: 38626093 PMCID: PMC11020485 DOI: 10.1371/journal.pone.0299215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 02/07/2024] [Indexed: 04/18/2024] Open
Abstract
Non-replicating adenovirus-based vectors have been broadly used for the development of prophylactic vaccines in humans and are licensed for COVID-19 and Ebola virus disease prevention. Adenovirus-based vectored vaccines encode for one or more disease specific transgenes with the aim to induce protective immunity against the target disease. The magnitude and duration of transgene expression of adenovirus 5- based vectors (human type C) in the host are key factors influencing antigen presentation and adaptive immune responses. Here we characterize the magnitude, duration, and organ biodistribution of transgene expression after single intramuscular administration of adenovirus 26-based vector vaccines in mice and evaluate the differences with adenovirus 5-based vector vaccine to understand if this is universally applicable across serotypes. We demonstrate a correlation between peak transgene expression early after adenovirus 26-based vaccination and transgene-specific cellular and humoral immune responses for a model antigen and SARS-CoV-2 spike protein, independent of innate immune activation. Notably, the memory immune response was similar in mice immunized with adenovirus 26-based vaccine and adenovirus 5-based vaccine, despite the latter inducing a higher peak of transgene expression early after immunization and a longer duration of transgene expression. Together these results provide further insights into the mode of action of adenovirus 26-based vector vaccines.
Collapse
Affiliation(s)
| | - Nadine Salisch
- Janssen Vaccines & Prevention B.V, Leiden, CN, The Netherlands
| | - Jan Serroyen
- Janssen Vaccines & Prevention B.V, Leiden, CN, The Netherlands
| | - Roland Zahn
- Janssen Vaccines & Prevention B.V, Leiden, CN, The Netherlands
| | - Selina Khan
- Janssen Vaccines & Prevention B.V, Leiden, CN, The Netherlands
| |
Collapse
|
2
|
Kapse B, Budev MM, Singer JP, Greenland JR. Immune aging: biological mechanisms, clinical symptoms, and management in lung transplant recipients. FRONTIERS IN TRANSPLANTATION 2024; 3:1356948. [PMID: 38993782 PMCID: PMC11235310 DOI: 10.3389/frtra.2024.1356948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 01/23/2024] [Indexed: 07/13/2024]
Abstract
While chronologic age can be precisely defined, clinical manifestations of advanced age occur in different ways and at different rates across individuals. The observed phenotype of advanced age likely reflects a superposition of several biological aging mechanisms which have gained increasing attention as the world contends with an aging population. Even within the immune system, there are multiple age-associated biological mechanisms at play, including telomere dysfunction, epigenetic dysregulation, immune senescence programs, and mitochondrial dysfunction. These biological mechanisms have associated clinical syndromes, such as telomere dysfunction leading to short telomere syndrome (STS), and optimal patient management may require recognition of biologically based aging syndromes. Within the clinical context of lung transplantation, select immune aging mechanisms are particularly pronounced. Indeed, STS is increasingly recognized as an indication for lung transplantation. At the same time, common aging phenotypes may be evoked by the stress of transplantation because lung allografts face a potent immune response, necessitating higher levels of immune suppression and associated toxicities, relative to other solid organs. Age-associated conditions exacerbated by lung transplant include bone marrow suppression, herpes viral infections, liver cirrhosis, hypogammaglobulinemia, frailty, and cancer risk. This review aims to dissect the molecular mechanisms of immune aging and describe their clinical manifestations in the context of lung transplantation. While these mechanisms are more likely to manifest in the context of lung transplantation, this mechanism-based approach to clinical syndromes of immune aging has broad relevance to geriatric medicine.
Collapse
Affiliation(s)
- Bhavya Kapse
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Marie M. Budev
- Department of Pulmonary Medicine, Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Jonathan P. Singer
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - John R. Greenland
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
- San Francisco VA Health Care System, Medicine, San Francisco, CA, United States
| |
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
|
Campbell E, Dobkin J, Osorio LJ, Kolloli A, Ramasamy S, Kumar R, Sant'Angelo DB, Subbian S, Denzin LK, Anderson S. A SARS-CoV-2 Vaccine Designed for Manufacturability Results in Unexpected Potency and Non-Waning Humoral Response. Vaccines (Basel) 2023; 11:vaccines11040832. [PMID: 37112744 PMCID: PMC10145385 DOI: 10.3390/vaccines11040832] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/03/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
The rapid development of several highly efficacious SARS-CoV-2 vaccines was an unprecedented scientific achievement that saved millions of lives. However, now that SARS-CoV-2 is transitioning to the endemic stage, there exists an unmet need for new vaccines that provide durable immunity and protection against variants and can be more easily manufactured and distributed. Here, we describe a novel protein component vaccine candidate, MT-001, based on a fragment of the SARS-CoV-2 spike protein that encompasses the receptor binding domain (RBD). Mice and hamsters immunized with a prime-boost regimen of MT-001 demonstrated extremely high anti-spike IgG titers, and remarkably this humoral response did not appreciably wane for up to 12 months following vaccination. Further, virus neutralization titers, including titers against variants such as Delta and Omicron BA.1, remained high without the requirement for subsequent boosting. MT-001 was designed for manufacturability and ease of distribution, and we demonstrate that these attributes are not inconsistent with a highly immunogenic vaccine that confers durable and broad immunity to SARS-CoV-2 and its emerging variants. These properties suggest MT-001 could be a valuable new addition to the toolbox of SARS-CoV-2 vaccines and other interventions to prevent infection and curtail additional morbidity and mortality from the ongoing worldwide pandemic.
Collapse
Affiliation(s)
- Elliot Campbell
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ 08854, USA
- Macrotope, Inc., Princeton, NJ 08540, USA
| | - Julie Dobkin
- Child Health Institute of New Jersey, Department of Pediatrics and Pharmacology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
- Graduate School of Biomedical Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Louis J Osorio
- Child Health Institute of New Jersey, Department of Pediatrics and Pharmacology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Afsal Kolloli
- Public Health Research Institute (PHRI), New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Santhamani Ramasamy
- Public Health Research Institute (PHRI), New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Ranjeet Kumar
- Public Health Research Institute (PHRI), New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Derek B Sant'Angelo
- Child Health Institute of New Jersey, Department of Pediatrics and Pharmacology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Selvakumar Subbian
- Public Health Research Institute (PHRI), New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Lisa K Denzin
- Child Health Institute of New Jersey, Department of Pediatrics and Pharmacology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Stephen Anderson
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ 08854, USA
- Macrotope, Inc., Princeton, NJ 08540, USA
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
5
|
Shimada M, Wang H, Ichino M, Ura T, Mizuki N, Okuda K. Biodistribution and immunity of adenovirus 5/35 and modified vaccinia Ankara vector vaccines against human immunodeficiency virus 1 clade C. Gene Ther 2022; 29:636-642. [PMID: 34987192 DOI: 10.1038/s41434-021-00308-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/06/2021] [Accepted: 11/23/2021] [Indexed: 01/09/2023]
Abstract
Previously, we developed a chimeric adenovirus type 5 with type 35 fiber (Ad5/35), which has high tropism to dendritic cells and low hepatoxicity. For further clinical use, we constructed two recombinant vectors expressing human immunodeficiency virus 1 (HIV-1) clade C gag (Ad5/35-Cgag and MVA-Cgag). The biodistribution of the two viral vectors in a mouse model and immunity in monkeys were assessed. The mice received a single intramuscular injection with the vectors alone. The gag gene in the tissues were periodically detected using a real-time quantitative polymerase chain reaction. The distribution of Ad5/35 was also detected using an in vivo imaging system, followed by luciferase-expressing Ad5/35 administration. We found that Ad5/35-Cgag DNA and luciferase activity were detectable until 8 weeks post-administration, whereas MVA-Cgag was undetectable 72 h post-administration. Furthermore, viral administration did not increase serum aspartate aminotransferase and alanine aminotransferase levels in either mouse or monkey models. Moreover, intramuscular administration of Ad5/35-Cgag induced the gag-specific antibody level and IFNγ-secreting PBMCs, the boost with MVA-Cgag further increased the responses and lasted more than 20 weeks from the initial administration. These data demonstrate that Ad5/35 and MVA vectors are safe for in vivo use, and prime-boost with Ad5/35-MVA vaccines is suitable for clinical use against HIV-1 clade C.
Collapse
Affiliation(s)
- Masaru Shimada
- Department of Molecular Biodefense Research, Yokohama City University, Yokohama, 2360004, Japan.
| | - Haibin Wang
- BioRay Pharmaceutical Co., Ltd., Taizhou, Zhejiang, 318000, China
| | - Motohide Ichino
- Department of Immunology, Yokohama City University, Yokohama, 2360004, Japan
| | - Takehiro Ura
- Department of Ophthalmology and Visual Science, Yokohama City University, Yokohama, 2360004, Japan
| | - Nobuhisa Mizuki
- Department of Ophthalmology and Visual Science, Yokohama City University, Yokohama, 2360004, Japan
| | - Kenji Okuda
- Department of Molecular Biodefense Research, Yokohama City University, Yokohama, 2360004, Japan.,Okuda Vaccine Research Institute, Yokohama, 2350045, Japan.,Yokohama City University, Yokohama, 2360004, Japan
| |
Collapse
|
6
|
Padron-Regalado E, Ulaszewska M, Douglas AD, Hill AVS, Spencer AJ. STING-pathway modulation to enhance the immunogenicity of adenoviral-vectored vaccines. Sci Rep 2022; 12:14464. [PMID: 36002507 PMCID: PMC9401198 DOI: 10.1038/s41598-022-18750-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 08/18/2022] [Indexed: 11/09/2022] Open
Abstract
Traditional chemical adjuvants remain a practical means of enhancing the immunogenicity of vaccines. Nevertheless, it is recognized that increasing the immunogenicity of viral vectors is challenging. Recently, STING ligands have been shown to enhance the efficacy of different vaccine platforms, but their affectivity on viral-vectored vaccination has not been fully assessed. In this study we used a multi-pronged approach to shed light on the immunological properties and potential mechanisms of action of this type of adjuvant and focused our study on replication-deficient human adenovirus serotype 5 (AdHu5). When the STING ligand 2'3'-cGAMP was mixed with AdHu5, the adjuvant enhanced anti-vector immune responses while decreasing the transgene-specific CD8+ T cell response. Studies employing STING-knockout mice and a 2'3'-cGAMP inactive analogue confirmed the aforementioned effects were STING dependent. In vitro assays demonstrated 2'3'-cGAMP induced the production of IFN-β which in turn negatively affected AdHu5 transgene expression and CD8+ T cell immunogenicity. In an effort to overcome the negative impact of early 2'3'-cGAMP signaling on AdHu5 transgene immunogenicity, we generated a bicistronic vector encoding the 2'3'-cGAMP together with a model antigen. Intracellular production of 2'3'-cGAMP after AdHu5 infection was able to enhance transgene-specific CD8+ T cell immunogenicity, although not to a level that would warrant progression of this adjuvant to clinical assessment. This work highlights the importance of timing of 2'3'-cGAMP administration when assessing its adjuvant capacity with different vaccine modalities.
Collapse
Affiliation(s)
- Eriko Padron-Regalado
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Marta Ulaszewska
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Alexander D Douglas
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Adrian V S Hill
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Alexandra J Spencer
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
7
|
McCann N, O'Connor D, Lambe T, Pollard AJ. Viral vector vaccines. Curr Opin Immunol 2022; 77:102210. [PMID: 35643023 PMCID: PMC9612401 DOI: 10.1016/j.coi.2022.102210] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/13/2022] [Accepted: 04/15/2022] [Indexed: 01/06/2023]
Abstract
Over the past two years, the SARS-CoV-2 pandemic has highlighted the impact that emerging pathogens can have on global health. The development of new and effective vaccine technologies is vital in the fight against such threats. Viral vectors are a relatively new vaccine platform that relies on recombinant viruses to deliver selected immunogens into the host. In response to the SARS-CoV-2 pandemic, the development and subsequent rollout of adenoviral vector vaccines has shown the utility, impact, scalability and efficacy of this platform. Shown to elicit strong cellular and humoral immune responses in diverse populations, these vaccine vectors will be an important approach against infectious diseases in the future.
Collapse
Affiliation(s)
- Naina McCann
- Oxford Vaccine Group, Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Headington, Oxford OX3 7LE, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| | - Daniel O'Connor
- Oxford Vaccine Group, Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Headington, Oxford OX3 7LE, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Teresa Lambe
- Oxford Vaccine Group, Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Headington, Oxford OX3 7LE, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Headington, Oxford OX3 7LE, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
8
|
Michiels Y, Houhou-Fidouh N, Collin G, Berger J, Kohli E. Humoral Response Induced by Prime-Boost Vaccination with the ChAdOx1 nCoV-19 and mRNA BNT162b2 Vaccines in a Teriflunomide-Treated Multiple Sclerosis Patient. Vaccines (Basel) 2021; 9:vaccines9101140. [PMID: 34696248 PMCID: PMC8540857 DOI: 10.3390/vaccines9101140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 09/23/2021] [Accepted: 10/02/2021] [Indexed: 11/16/2022] Open
Abstract
Patients with multiple sclerosis (MS) are treated with drugs that may impact immune responses to SARS-CoV-2 vaccination. Evaluation of “prime-boost” (heterologous) vaccination regimens including a first administration of a viral vector-based vaccine and a second one of an mRNA-based vaccine in such patients has not yet been completed. Here, we present the anti-spike protein S humoral response, including the neutralizing antibody response, in a 54-year-old MS patient who had been treated with teriflunomide for the past 2 years and who received a heterologous ChAdOx1 nCoV-19/ BNT162b2 vaccination regimen. The results showed a very strong anti-S IgG response and a good neutralizing antibody response. These results show that teriflunomide did not prevent the development of a satisfactory humoral response in this MS patient after vaccination with a ChAdOx1 nCoV-19/ BNT162b2 prime-boost protocol.
Collapse
Affiliation(s)
- Yves Michiels
- Community Pharmacy, Center for Primary Care and Public Health (Unisanté) Lausanne 1011, University of Lausanne, 1015 Lausanne, Switzerland;
- Pharmacie Michiels, Research Department, 21600 Longvic, France
- Correspondence:
| | - Nadhira Houhou-Fidouh
- Laboratoire de Virologie, Hôpital Bichat, Sorbonne Paris Cité, Université Paris Diderot, AP-HP, 75018 Paris, France; (N.H.-F.); (G.C.)
| | - Gilles Collin
- Laboratoire de Virologie, Hôpital Bichat, Sorbonne Paris Cité, Université Paris Diderot, AP-HP, 75018 Paris, France; (N.H.-F.); (G.C.)
| | - Jérôme Berger
- Community Pharmacy, Center for Primary Care and Public Health (Unisanté) Lausanne 1011, University of Lausanne, 1015 Lausanne, Switzerland;
- School of Pharmaceutical Sciences, University of Geneva, 1205 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Lausanne, 1015 Lausanne, Switzerland
| | - Evelyne Kohli
- UMR INSERM/uB/AGROSUP 1231, Team 3 HSP-Pathies, Labellisée Ligue Nationale Contre le Cancer and Laboratoire d’Excellence LipSTIC, 21000 Dijon, France;
- UFR des Sciences de Santé, Université de Bourgogne, 21000 Dijon, France
- Univesity Hospital, 21000 Dijon, France
| |
Collapse
|
9
|
Li Q, Wang J, Tang Y, Lu H. Next-generation COVID-19 vaccines: Opportunities for vaccine development and challenges in tackling COVID-19. Drug Discov Ther 2021; 15:118-123. [PMID: 34234059 DOI: 10.5582/ddt.2021.01058] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The ongoing COVID-19 pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become a global threat. Although non-pharmaceutical interventions have been rigorously and widely implemented, living conditions caused by the pandemic will last until highly effective vaccines are successfully improved and globally administered. Several first-generation COVID-19 vaccines were approved at the end of 2020. However, the COVID-19 pandemic is persisting worldwide. To be clear, the efficiency and the coverage of current vaccines are insufficient, but newly emerging and rapidly spreading variants are the most pressing concern. A second-generation COVID-19 vaccine worth mentioning, NVX-CoV2373, has demonstrated 90% overall efficacy as well as a high level of efficacy against circulating variants in Phase 3 clinical trials. Currently, NVX-CoV2373 is the only vaccine that has proven successful against variants during Phase 3/4 trials. Therefore, developing the next generation of vaccines is a promising strategy to ultimately prevail against SARS-CoV-2. This review provides up-to-date information on COVID-19 vaccines in terms of their efficacy and new platforms and the progression of COVID-19 vaccination. Moreover, this review also summarizes the efficacy of approved COVID-19 vaccines against variants. Lastly, this review highlights the global challenges for COVID-19 vaccines in development and vaccination, and it discusses opportunities for development of future COVID-19 vaccines and vaccination coverage.
Collapse
Affiliation(s)
- Qian Li
- Department of Infectious Diseases, Shanghai Public Health Clinical Center, Shanghai, China
| | - Jun Wang
- Center of Clinical Laboratory, The Fifth People's Hospital of Wuxi, Jiangnan University, Wuxi, China
| | - Yang Tang
- Department of Infectious Diseases, Shanghai Public Health Clinical Center, Shanghai, China
| | - Hongzhou Lu
- Department of Infectious Diseases, Shanghai Public Health Clinical Center, Shanghai, China
| |
Collapse
|
10
|
Larson C, Oronsky B, Goyal S, Ray C, Hedjran F, Hammond TC, Kesari S, Caroen S, Lybeck M, Dobalian VE, Oronsky A, Reid T. COVID-19 and cancer: A guide with suggested COVID-19 rule-out criteria to support clinical decision-making. Biochim Biophys Acta Rev Cancer 2020; 1874:188412. [PMID: 32827582 PMCID: PMC7438345 DOI: 10.1016/j.bbcan.2020.188412] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 12/21/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a highly contagious zoonotic pathogen that has exacted heavy public health, social and economic tolls. In February 2020, the World Health Organization acronymed the disease caused by SARS-CoV-2 as COVID-19, for coronavirus disease 2019. The number of confirmed COVID-19 infections, which has been detected in at least 103 countries, has reached 1,970,225 worldwide as of April 14, 2020 with 124,544 deaths, according to the U.S. Centers for Disease Control and Prevention (CDC). Many cases of COVID-19 resolve quickly. However, the disease, which, like other respiratory pathogens that cause common cold symptoms is believed to be transmitted through respiratory droplets. Infection with COVID-19 can also lead to significant morbidity and death; this is particularly the case for cancer patients. Moreover, because the signs and symptoms of COVID-19 are easily misattributed to the sequelae of cancer itself, such as pulmonary embolism, or its treatment, such as nausea and diarrhea, diagnosis may be delayed or missed. Potential COVID-19 rule out criteria, based on the Wells' criteria for pulmonary embolism, another protean disease entity, are provided as a decision-making aid. This review summarizes the current understanding of the transmission, clinical presentation, diagnosis and differential diagnosis, pathogenesis, rationale to treat the cancer or not, treatment and prevention of COVID-19 with an emphasis on implications in cancer.
Collapse
Affiliation(s)
| | | | - Sharad Goyal
- George Washington University, Washington, DC, USA
| | - Carolyn Ray
- St. Francis Hospital and Medical Center, Hartford, CT, USA
| | | | - Terese C Hammond
- John Wayne Cancer Institute at Providence St. John's Health, Santa Monica, CA, USA
| | - Santosh Kesari
- John Wayne Cancer Institute at Providence St. John's Health, Santa Monica, CA, USA
| | | | | | | | | | | |
Collapse
|
11
|
Rawat K, Kumari P, Saha L. COVID-19 vaccine: A recent update in pipeline vaccines, their design and development strategies. Eur J Pharmacol 2020; 892:173751. [PMID: 33245898 PMCID: PMC7685956 DOI: 10.1016/j.ejphar.2020.173751] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 12/20/2022]
Abstract
Coronavirus Disease 2019 named as COVID-19 imposing a huge burden on public health as well as global economies, is caused by a new strain of betacoronavirus named as SARS-CoV-2. The high transmission rate of the virus has resulted in current havoc which highlights the need for a fast and effective approach either to prevent or treat the deadly infection. Development of vaccines can be the most prominent approach to prevent the virus to cause COVID-19 and hence will play a vital role in controlling the spread of the virus and reducing mortality. The virus uses its spike proteins for entering into the host by interacting with a specific receptor called angiotensin converting enzyme-2 (ACE2) present on the surface of alveolar cells in the lungs. Researchers all over the world are targeting the spike protein for the development of potential vaccines. Here, we discuss the immunopathological basis of vaccine designing that can be approached for vaccine development against SARS-CoV-2 infection and different platforms that are being used for vaccine development. We believe this review will increase our understanding of the vaccine designing against SARS-CoV-2 and subsequently contribute to the control of SARS-CoV-2 infections. Also, it gives an insight into the current status of vaccine development and associated outcomes reported at different phases of trial. Either the S protein of the SARS-CoV-2 or the whole pathogen is targeted at the development of vaccines against COVID-19. Lymphocytopenia and Cytokine storm are two major manifestations associated with innate immune response generated against COVID-19. There are 44 vaccine candidates in clinical phase and 154 in preclinical phase of vaccine development. The major hurdle in establishing vaccine's efficacy being its effectiveness and safety at each step among diverse population.
Collapse
Affiliation(s)
- Kajal Rawat
- Department of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), 4th Floor, Research Block B, Chandigarh, 160012, India
| | - Puja Kumari
- Department of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), 4th Floor, Research Block B, Chandigarh, 160012, India
| | - Lekha Saha
- Department of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), 4th Floor, Research Block B, Chandigarh, 160012, India.
| |
Collapse
|
12
|
Lofano G, Mallett CP, Bertholet S, O’Hagan DT. Technological approaches to streamline vaccination schedules, progressing towards single-dose vaccines. NPJ Vaccines 2020; 5:88. [PMID: 33024579 PMCID: PMC7501859 DOI: 10.1038/s41541-020-00238-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/25/2020] [Indexed: 12/21/2022] Open
Abstract
Vaccines represent the most successful medical intervention in history, with billions of lives saved. Although multiple doses of the same vaccine are typically required to reach an adequate level of protection, it would be advantageous to develop vaccines that induce protective immunity with fewer doses, ideally just one. Single-dose vaccines would be ideal to maximize vaccination coverage, help stakeholders to greatly reduce the costs associated with vaccination, and improve patient convenience. Here we describe past attempts to develop potent single dose vaccines and explore the reasons they failed. Then, we review key immunological mechanisms of the vaccine-specific immune responses, and how innovative technologies and approaches are guiding the preclinical and clinical development of potent single-dose vaccines. By modulating the spatio-temporal delivery of the vaccine components, by providing the appropriate stimuli to the innate immunity, and by designing better antigens, the new technologies and approaches leverage our current knowledge of the immune system and may synergize to enable the rational design of next-generation vaccination strategies. This review provides a rational perspective on the possible development of future single-dose vaccines.
Collapse
Affiliation(s)
- Giuseppe Lofano
- GSK, Slaoui Center for Vaccines Research, Rockville, MD 20850 USA
| | - Corey P. Mallett
- GSK, Slaoui Center for Vaccines Research, Rockville, MD 20850 USA
| | - Sylvie Bertholet
- GSK, Slaoui Center for Vaccines Research, Rockville, MD 20850 USA
| | - Derek T. O’Hagan
- GSK, Slaoui Center for Vaccines Research, Rockville, MD 20850 USA
| |
Collapse
|
13
|
Irvine DJ, Aung A, Silva M. Controlling timing and location in vaccines. Adv Drug Deliv Rev 2020; 158:91-115. [PMID: 32598970 PMCID: PMC7318960 DOI: 10.1016/j.addr.2020.06.019] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023]
Abstract
Vaccines are one of the most powerful technologies supporting public health. The adaptive immune response induced by immunization arises following appropriate activation and differentiation of T and B cells in lymph nodes. Among many parameters impacting the resulting immune response, the presence of antigen and inflammatory cues for an appropriate temporal duration within the lymph nodes, and further within appropriate subcompartments of the lymph nodes- the right timing and location- play a critical role in shaping cellular and humoral immunity. Here we review recent advances in our understanding of how vaccine kinetics and biodistribution impact adaptive immunity, and the underlying immunological mechanisms that govern these responses. We discuss emerging approaches to engineer these properties for future vaccines, with a focus on subunit vaccines.
Collapse
Affiliation(s)
- Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA 92037, USA; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| | - Aereas Aung
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Murillo Silva
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
14
|
Greenland JR, Michelow MD, Wang L, London MJ. COVID-19 Infection: Implications for Perioperative and Critical Care Physicians. Anesthesiology 2020; 132:1346-1361. [PMID: 32195698 PMCID: PMC7155909 DOI: 10.1097/aln.0000000000003303] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 03/10/2020] [Indexed: 01/08/2023]
Abstract
Healthcare systems worldwide are responding to Coronavirus Disease 2019 (COVID-19), an emerging infectious syndrome caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) virus. Patients with COVID-19 can progress from asymptomatic or mild illness to hypoxemic respiratory failure or multisystem organ failure, necessitating intubation and intensive care management. Healthcare providers, and particularly anesthesiologists, are at the frontline of this epidemic, and they need to be aware of the best available evidence to guide therapeutic management of patients with COVID-19 and to keep themselves safe while doing so. Here, the authors review COVID-19 pathogenesis, presentation, diagnosis, and potential therapeutics, with a focus on management of COVID-19-associated respiratory failure. The authors draw on literature from other viral epidemics, treatment of acute respiratory distress syndrome, and recent publications on COVID-19, as well as guidelines from major health organizations. This review provides a comprehensive summary of the evidence currently available to guide management of critically ill patients with COVID-19.
Collapse
Affiliation(s)
- John R Greenland
- From Pulmonary and Critical Care Medicine (J.R.G.) Anesthesia and Perioperative Care (M.D.M., M.J.L.), San Francisco Veterans Administration Health Care System, San Francisco, California Laboratory Medicine (L.W.) University of California, San Francisco, California (J.R.G., M.D.M., M.J.L.)
| | | | | | | |
Collapse
|
15
|
Kugler F, Drexler I, Protzer U, Hoffmann D, Moeini H. Generation of recombinant MVA-norovirus: a comparison study of bacterial artificial chromosome- and marker-based systems. Virol J 2019; 16:100. [PMID: 31399106 PMCID: PMC6688233 DOI: 10.1186/s12985-019-1212-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 08/05/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Recombinant Modified Vaccinia Virus Ankara has been employed as a safe and potent viral vector vaccine against infectious diseases and cancer. We generated recMVAs encoding norovirus GII.4 genotype capsid protein by using a marker-based approach and a BAC-based system. In the marker-based approach, the capsid gene together with a reporter gene was introduced into the MVA genome in DF-1 cells. Several rounds of plaque purification were carried out to get rid of the WT-MVA. In the BAC-based approach, recMVA-BAC was produced by en passant recombineering in E. coli. Subsequently, the recMVAs were rescued in DF-1 cells using a helper rabbit fibroma virus. The BAC backbone and the helper virus were eliminated by passaging in DF-1 cells. Biochemical characteristics of the recMVAs were studied. RESULTS We found the purification of the rare spontaneous recombinants time-consuming in the marker-based system. In contrast, the BAC-based system rapidly inserted the gene of interest in E. coli by en passant recombineering before virion production in DF-1 cells. The elimination of the reporter gene was found to be faster and more efficient in the BAC-based approach. With Western blotting and electron microscopy, we could prove successful capsid protein expression and proper virus-assembly, respectively. The MVA-BAC produced higher recombinant virus titers and infected DF-1 cells more efficiently. CONCLUSIONS Comparing both methods, we conclude that, in contrast to the tedious and time-consuming traditional method, the MVA-BAC system allows us to quickly generate high titer recMVAs.
Collapse
Affiliation(s)
- Franziska Kugler
- Institute of Virology, Faculty of Medicine, Technische Universität München, Munich, Germany
| | - Ingo Drexler
- Institute for Virology, Universitätklinikum Düsseldorf, Heinrich Heine Universität, Düsseldorf, Germany
| | - Ulrike Protzer
- Institute of Virology, Faculty of Medicine, Technische Universität München, Munich, Germany
| | - Dieter Hoffmann
- Institute of Virology, Faculty of Medicine, Technische Universität München, Munich, Germany.
| | - Hassan Moeini
- Institute of Virology, Faculty of Medicine, Technische Universität München, Munich, Germany.
| |
Collapse
|
16
|
Wang C, Dulal P, Zhou X, Xiang Z, Goharriz H, Banyard A, Green N, Brunner L, Ventura R, Collin N, Draper SJ, Hill AVS, Ashfield R, Fooks AR, Ertl HC, Douglas AD. A simian-adenovirus-vectored rabies vaccine suitable for thermostabilisation and clinical development for low-cost single-dose pre-exposure prophylaxis. PLoS Negl Trop Dis 2018; 12:e0006870. [PMID: 30372438 PMCID: PMC6224154 DOI: 10.1371/journal.pntd.0006870] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 11/08/2018] [Accepted: 09/24/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Estimates of current global rabies mortality range from 26,000 to 59,000 deaths per annum. Although pre-exposure prophylaxis using inactivated rabies virus vaccines (IRVs) is effective, it requires two to three doses and is regarded as being too expensive and impractical for inclusion in routine childhood immunization programmes. METHODOLOGY/ PRINCIPAL FINDINGS Here we report the development of a simian-adenovirus-vectored rabies vaccine intended to enable cost-effective population-wide pre-exposure prophylaxis against rabies. ChAdOx2 RabG uses the chimpanzee adenovirus serotype 68 (AdC68) backbone previously shown to achieve pre-exposure protection against rabies in non-human primates. ChAdOx2 differs from AdC68 in that it contains the human adenovirus serotype 5 (AdHu5) E4 orf6/7 region in place of the AdC68 equivalents, enhancing ease of manufacturing in cell lines which provide AdHu5 E1 proteins in trans. We show that immunogenicity of ChAdOx2 RabG in mice is comparable to that of AdC68 RabG and other adenovirus serotypes expressing rabies virus glycoprotein. High titers of rabies virus neutralizing antibody (VNA) are elicited after a single dose. The relationship between levels of VNA activity and rabies virus glycoprotein monomer-binding antibody differs after immunization with adenovirus-vectored vaccines and IRV vaccines, suggesting routes to further enhancement of the efficacy of the adenovirus-vectored candidates. We also demonstrate that ChAdOx2 RabG can be thermostabilised using a low-cost method suitable for clinical bio-manufacture and ambient-temperature distribution in tropical climates. Finally, we show that a dose-sparing effect can be achieved by formulating ChAdOx2 RabG with a simple chemical adjuvant. This approach could lower the cost of ChAdOx2 RabG and other adenovirus-vectored vaccines. CONCLUSIONS/ SIGNIFICANCE ChAdOx2 RabG may prove to be a useful tool to reduce the human rabies death toll. We have secured funding for Good Manufacturing Practice- compliant bio-manufacture and Phase I clinical trial of this candidate.
Collapse
Affiliation(s)
- Chuan Wang
- Jenner Institute, University of Oxford, Wellcome Trust Centre for Human Genetics, Oxford, United Kingdom
| | - Pawan Dulal
- Jenner Institute, University of Oxford, Wellcome Trust Centre for Human Genetics, Oxford, United Kingdom
| | - Xiangyang Zhou
- Wistar Institute of Anatomy & Biology, Philadelphia, Pennsylvania, United States of America
| | - Zhiquan Xiang
- Wistar Institute of Anatomy & Biology, Philadelphia, Pennsylvania, United States of America
| | - Hooman Goharriz
- Animal and Plant Health Agency (APHA), Wildlife Zoonoses and Vector-borne Diseases Research Group, New Haw, Surrey, United Kingdom
| | - Ashley Banyard
- Animal and Plant Health Agency (APHA), Wildlife Zoonoses and Vector-borne Diseases Research Group, New Haw, Surrey, United Kingdom
| | - Nicky Green
- Clinical Biomanufacturing Facility, University of Oxford, Oxford, United Kingdom
| | - Livia Brunner
- Vaccine Formulation Laboratory, University of Lausanne, Epalinges, Switzerland
| | - Roland Ventura
- Vaccine Formulation Laboratory, University of Lausanne, Epalinges, Switzerland
| | - Nicolas Collin
- Vaccine Formulation Laboratory, University of Lausanne, Epalinges, Switzerland
| | - Simon J. Draper
- Jenner Institute, University of Oxford, Wellcome Trust Centre for Human Genetics, Oxford, United Kingdom
| | - Adrian V. S. Hill
- Jenner Institute, University of Oxford, Wellcome Trust Centre for Human Genetics, Oxford, United Kingdom
| | - Rebecca Ashfield
- Jenner Institute, University of Oxford, Wellcome Trust Centre for Human Genetics, Oxford, United Kingdom
| | - Anthony R. Fooks
- Animal and Plant Health Agency (APHA), Wildlife Zoonoses and Vector-borne Diseases Research Group, New Haw, Surrey, United Kingdom
| | - Hildegund C. Ertl
- Wistar Institute of Anatomy & Biology, Philadelphia, Pennsylvania, United States of America
| | - Alexander D. Douglas
- Jenner Institute, University of Oxford, Wellcome Trust Centre for Human Genetics, Oxford, United Kingdom
| |
Collapse
|
17
|
Alharbi NK. Poxviral promoters for improving the immunogenicity of MVA delivered vaccines. Hum Vaccin Immunother 2018; 15:203-209. [PMID: 30148692 DOI: 10.1080/21645515.2018.1513439] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Modified vaccinia virus Ankara (MVA) is a replication-deficient poxvirus, attenuated in chick embryo fibroblast primary cells. It has been utilised as a viral vector to develop many vaccines against cancer and infectious diseases such as malaria, HIV/AIDS, influenza, and tuberculosis, MERS-CoV, and Ebola virus infection. There is accumulating data from many preclinical and clinical studies that highlights the excellent safety and immunogenicity of MVA. However, due to the complex nature of many pathogens and their pathogenicity, MVA vectored vaccine candidates need to be optimised to improve their immunogenicity. One of the main approaches to improve MVA immunogenicity focuses on optimising poxviral promoters that drive recombinant vaccine antigens, encoded within recombinant MVA vector genome. A number of promoters were described or optimised to improve the development of MVA based vaccines such as p7.5, pF11, and mH5 promoters. This review focuses on poxviral promoters, their optimisation, genetic stability, and clinical use.
Collapse
Affiliation(s)
- Naif Khalaf Alharbi
- a Infectious Disease Research Department , King Abdullah International Medical Research Center (KAIMRC) , Riyadh , Saudi Arabia
| |
Collapse
|
18
|
Kumar A, Meldgaard TS, Bertholet S. Novel Platforms for the Development of a Universal Influenza Vaccine. Front Immunol 2018; 9:600. [PMID: 29628926 PMCID: PMC5877485 DOI: 10.3389/fimmu.2018.00600] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 03/09/2018] [Indexed: 12/19/2022] Open
Abstract
Despite advancements in immunotherapeutic approaches, influenza continues to cause severe illness, particularly among immunocompromised individuals, young children, and elderly adults. Vaccination is the most effective way to reduce rates of morbidity and mortality caused by influenza viruses. Frequent genetic shift and drift among influenza-virus strains with the resultant disparity between circulating and vaccine virus strains limits the effectiveness of the available conventional influenza vaccines. One approach to overcome this limitation is to develop a universal influenza vaccine that could provide protection against all subtypes of influenza viruses. Moreover, the development of a novel or improved universal influenza vaccines may be greatly facilitated by new technologies including virus-like particles, T-cell-inducing peptides and recombinant proteins, synthetic viruses, broadly neutralizing antibodies, and nucleic acid-based vaccines. This review discusses recent scientific advances in the development of next-generation universal influenza vaccines.
Collapse
Affiliation(s)
- Arun Kumar
- GSK, Research and Development Center, Siena, Italy.,Linköping University, Linköping, Sweden
| | - Trine Sundebo Meldgaard
- GSK, Research and Development Center, Siena, Italy.,DTU Nanotech, Technical University of Denmark, Copenhagen, Denmark
| | - Sylvie Bertholet
- GSK, Research and Development Center, Siena, Italy.,GSK, Research and Development Center, Rockville, MD, United States
| |
Collapse
|
19
|
Larocca RA, Provine NM, Aid M, Iampietro MJ, Borducchi EN, Badamchi-Zadeh A, Abbink P, Ng'ang'a D, Bricault CA, Blass E, Penaloza-MacMaster P, Stephenson KE, Barouch DH. Adenovirus serotype 5 vaccine vectors trigger IL-27-dependent inhibitory CD4 + T cell responses that impair CD8 + T cell function. Sci Immunol 2016; 1. [PMID: 28239679 DOI: 10.1126/sciimmunol.aaf7643] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Adenovirus serotype 5 (Ad5) vaccine vectors elicit robust CD8+ T cell responses, but these responses typically exhibit a partially exhausted phenotype. However, the immunologic mechanism by which Ad5 vectors induce dysfunctional CD8+ T cells has not previously been elucidated. Here we demonstrate that, following immunization of B6 mice, Ad5 vectors elicit antigen-specific IL-10+CD4+ T cells with a distinct transcriptional profile in a dose-dependent fashion. In rhesus monkeys, we similarly observed upregulated expression of IL-10 and PD-1 by CD4+ T cells following Ad5 vaccination. These cells markedly suppressed vaccine-elicited CD8+ T cell responses in vivo and IL-10 blockade increased the frequency and functionality of antigen-specific CD8+ T cells as well as improved protective efficacy against challenge with recombinant Listeria monocytogenes. Moreover, induction of these inhibitory IL-10+CD4+ T cells correlated with IL-27 expression and IL-27 blockade substantially improved CD4+ T cell functionality. These data highlight a role for IL-27 in the induction of inhibitory IL-10+CD4+ T cells, which suppress CD8+ T cell magnitude and function following Ad5 vector immunization. A deeper understanding of the cytokine networks and transcriptional profiles induced by vaccine vectors should lead to strategies to improve the immunogenicity and protective efficacy of viral vector-based vaccines.
Collapse
Affiliation(s)
- Rafael A Larocca
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Nicholas M Provine
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Malika Aid
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - M Justin Iampietro
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Erica N Borducchi
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Alexander Badamchi-Zadeh
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Peter Abbink
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - David Ng'ang'a
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Christine A Bricault
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Eryn Blass
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Pablo Penaloza-MacMaster
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Microbiology and Immunology, Northwestern University, Chicago, IL 66611, USA
| | - Kathryn E Stephenson
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| |
Collapse
|
20
|
Wang C, Hart M, Chui C, Ajuogu A, Brian IJ, de Cassan SC, Borrow P, Draper SJ, Douglas AD. Germinal Center B Cell and T Follicular Helper Cell Responses to Viral Vector and Protein-in-Adjuvant Vaccines. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:1242-51. [PMID: 27412417 PMCID: PMC4974488 DOI: 10.4049/jimmunol.1502472] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 06/09/2016] [Indexed: 11/19/2022]
Abstract
There is great interest in the development of Ab-inducing subunit vaccines targeting infections, including HIV, malaria, and Ebola. We previously reported that adenovirus vectored vaccines are potent in priming Ab responses, but uncertainty remains regarding the optimal approach for induction of humoral immune responses. In this study, using OVA as a model Ag, we assessed the magnitude of the primary and anamnestic Ag-specific IgG responses of mice to four clinically relevant vaccine formulations: replication-deficient adenovirus; modified vaccinia Ankara (a poxvirus); protein with alum; and protein in the squalene oil-in-water adjuvant Addavax. We then used flow cytometric assays capable of measuring total and Ag-specific germinal center (GC) B cell and follicular Th cell responses to compare the induction of these responses by the different formulations. We report that adenovirus vectored vaccines induce Ag insert-specific GC B cell and Ab responses of a magnitude comparable to those induced by a potent protein/squalene oil-in-water formulation whereas-despite a robust overall GC response-the insert-specific GC B cell and Ab responses induced by modified vaccinia Ankara were extremely weak. Ag-specific follicular Th cell responses to adenovirus vectored vaccines exceeded those induced by other platforms at day 7 after immunization. We found little evidence that innate immune activation by adenovirus may act as an adjuvant in such a manner that the humoral response to a recombinant protein may be enhanced by coadministering with an adenovirus lacking a transgene of interest. Overall, these studies provide further support for the use of replication-deficient adenoviruses to induce humoral responses.
Collapse
Affiliation(s)
- Chuan Wang
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, United Kingdom; and
| | - Matthew Hart
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, United Kingdom; and
| | - Cecilia Chui
- Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, United Kingdom
| | - Augustine Ajuogu
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, United Kingdom; and
| | - Iona J Brian
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, United Kingdom; and
| | - Simone C de Cassan
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, United Kingdom; and
| | - Persephone Borrow
- Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, United Kingdom
| | - Simon J Draper
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, United Kingdom; and
| | - Alexander D Douglas
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, United Kingdom; and
| |
Collapse
|
21
|
Rady HF, Dai G, Huang W, Shellito JE, Ramsay AJ. Flagellin Encoded in Gene-Based Vector Vaccines Is a Route-Dependent Immune Adjuvant. PLoS One 2016; 11:e0148701. [PMID: 26844553 PMCID: PMC4742079 DOI: 10.1371/journal.pone.0148701] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 01/20/2016] [Indexed: 12/21/2022] Open
Abstract
Flagellin has been tested as a protein-based vaccine adjuvant, with the majority of studies focused on antibody responses. Here, we evaluated the adjuvant activity of flagellin for both cellular and humoral immune responses in BALB/c mice in the setting of gene-based immunization, and have made several novel observations. DNA vaccines and adenovirus (Ad) vectors were engineered to encode mycobacterial protein Ag85B, with or without flagellin of Salmonella typhimurium (FliC). DNA-encoded flagellin given IM enhanced splenic CD4+ and CD8+ T cell responses to co-expressed vaccine antigen, including memory responses. Boosting either IM or intranasally with Ad vectors expressing Ag85B without flagellin led to durable enhancement of Ag85B-specific antibody and CD4+ and CD8+ T cell responses in both spleen and pulmonary tissues, correlating with significantly improved protection against challenge with pathogenic aerosolized M. tuberculosis. However, inclusion of flagellin in both DNA prime and Ad booster vaccines induced localized pulmonary inflammation and transient weight loss, with route-dependent effects on vaccine-induced T cell immunity. The latter included marked reductions in levels of mucosal CD4+ and CD8+ T cell responses following IM DNA/IN Ad mucosal prime-boosting, although antibody responses were not diminished. These findings indicate that flagellin has differential and route-dependent adjuvant activity when included as a component of systemic or mucosally-delivered gene-based prime-boost immunization. Clear adjuvant activity for both T and B cell responses was observed when flagellin was included in the DNA priming vaccine, but side effects occurred when given in an Ad boosting vector, particularly via the pulmonary route.
Collapse
Affiliation(s)
- Hamada F. Rady
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
- Louisiana Vaccine Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Guixiang Dai
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
- Louisiana Vaccine Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Weitao Huang
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Judd E. Shellito
- Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
- Louisiana Vaccine Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Alistair J. Ramsay
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
- Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
- Louisiana Vaccine Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
22
|
García-Arriaza J, Esteban M. Enhancing poxvirus vectors vaccine immunogenicity. Hum Vaccin Immunother 2015; 10:2235-44. [PMID: 25424927 DOI: 10.4161/hv.28974] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Attenuated recombinant poxvirus vectors expressing heterologous antigens from pathogens are currently at various stages in clinical trials with the aim to establish their efficacy. This is because these vectors have shown excellent safety profiles, significant immunogenicity against foreign expressed antigens and are able to induce protective immune responses. In view of the limited efficacy triggered by some poxvirus strains used in clinical trials (i.e, ALVAC in the RV144 phase III clinical trial for HIV), and of the restrictive replication capacity of the highly attenuated vectors like MVA and NYVAC, there is a consensus that further improvements of these vectors should be pursuit. In this review we considered several strategies that are currently being implemented, as well as new approaches, to improve the immunogenicity of the poxvirus vectors. This includes heterologous prime/boost protocols, use of co-stimulatory molecules, deletion of viral immunomodulatory genes still present in the poxvirus genome, enhancing virus promoter strength, enhancing vector replication capacity, optimizing expression of foreign heterologous sequences, and the combined use of adjuvants. An optimized poxvirus vector triggering long-lasting immunity with a high protective efficacy against a selective disease should be sought.
Collapse
Affiliation(s)
- Juan García-Arriaza
- a Department of Molecular and Cellular Biology; Centro Nacional de Biotecnología; Consejo Superior de Investigaciones Científicas (CSIC); Madrid, Spain
| | | |
Collapse
|
23
|
Kinnear E, Caproni LJ, Tregoning JS. A Comparison of Red Fluorescent Proteins to Model DNA Vaccine Expression by Whole Animal In Vivo Imaging. PLoS One 2015; 10:e0130375. [PMID: 26091084 PMCID: PMC4475043 DOI: 10.1371/journal.pone.0130375] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 05/19/2015] [Indexed: 11/19/2022] Open
Abstract
DNA vaccines can be manufactured cheaply, easily and rapidly and have performed well in pre-clinical animal studies. However, clinical trials have so far been disappointing, failing to evoke a strong immune response, possibly due to poor antigen expression. To improve antigen expression, improved technology to monitor DNA vaccine transfection efficiency is required. In the current study, we compared plasmid encoded tdTomato, mCherry, Katushka, tdKatushka2 and luciferase as reporter proteins for whole animal in vivo imaging. The intramuscular, subcutaneous and tattooing routes were compared and electroporation was used to enhance expression. We observed that overall, fluorescent proteins were not a good tool to assess expression from DNA plasmids, with a highly heterogeneous response between animals. Of the proteins used, intramuscular delivery of DNA encoding either tdTomato or luciferase gave the clearest signal, with some Katushka and tdKatushka2 signal observed. Subcutaneous delivery was weakly visible and nothing was observed following DNA tattooing. DNA encoding haemagglutinin was used to determine whether immune responses mirrored visible expression levels. A protective immune response against H1N1 influenza was induced by all routes, even after a single dose of DNA, though qualitative differences were observed, with tattooing leading to high antibody responses and subcutaneous DNA leading to high CD8 responses. We conclude that of the reporter proteins used, expression from DNA plasmids can best be assessed using tdTomato or luciferase. But, the disconnect between visible expression level and immunogenicity suggests that in vivo whole animal imaging of fluorescent proteins has limited utility for predicting DNA vaccine efficacy.
Collapse
Affiliation(s)
- Ekaterina Kinnear
- Mucosal Infection & Immunity Group, Section of Virology, Imperial College London, St Mary’s Campus, London, United Kingdom
| | - Lisa J. Caproni
- Touchlight Genetics Ltd., Leatherhead, Surrey, United Kingdom
| | - John S. Tregoning
- Mucosal Infection & Immunity Group, Section of Virology, Imperial College London, St Mary’s Campus, London, United Kingdom
| |
Collapse
|
24
|
Alharbi NK, Spencer AJ, Hill AVS, Gilbert SC. Deletion of Fifteen Open Reading Frames from Modified Vaccinia Virus Ankara Fails to Improve Immunogenicity. PLoS One 2015; 10:e0128626. [PMID: 26053118 PMCID: PMC4459983 DOI: 10.1371/journal.pone.0128626] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 04/29/2015] [Indexed: 12/25/2022] Open
Abstract
Modified vaccinia virus Ankara (MVA) is a highly attenuated strain of vaccinia virus, which has been used as a recombinant vaccine vector in many vaccine development programmes. The loss of many immunosuppressive and host-range genes resulted in a safe and immunogenic vaccine vector. However it still retains some immunomodulatory genes that may reduce MVA immunogenicity. Earlier reports demonstrated that the deletion of the A41L, B15R, C6L, or C12L open reading frames (ORFs) enhanced cellular immune responses in recombinant MVA (rMVA) by up to 2-fold. However, previously, we showed that deletion of the C12L, A44L, A46R, B7R, or B15R ORFs from rMVA, using MVA-BAC recombineering technology, did not enhance rMVA immunogenicity at either peak or memory cellular immune responses. Here, we extend our previous study to examine the effect of deleting clusters of genes on rMVA cellular immunogenicity. Two clusters of fifteen genes were deleted in one rMVA mutant that encodes either the 85A antigen of Mycobacterium tuberculosis or an immunodominant H2-Kd-restricted murine malaria epitope (pb9). The deletion mutants were tested in prime only or prime and boost vaccination regimens. The responses showed no improved peak or memory CD8+ T cell frequencies. Our results suggest that the reported small increases in MVA deletion mutants could not be replicated with different antigens, or epitopes. Therefore, the gene deletion strategy may not be taken as a generic approach for improving the immunogenicity of MVA-based vaccines, and should be carefully assessed for every individual recombinant antigen.
Collapse
Affiliation(s)
- Naif Khalaf Alharbi
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, United Kingdom
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | | | - Adrian V. S. Hill
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, United Kingdom
| | - Sarah C. Gilbert
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, United Kingdom
| |
Collapse
|
25
|
Giles C, Vanniasinkam T, Ndi S, Barton MD. Rhodococcus equi (Prescottella equi)vaccines; the future of vaccine development. Equine Vet J 2014; 47:510-8. [DOI: 10.1111/evj.12310] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 06/12/2014] [Indexed: 12/29/2022]
Affiliation(s)
- C. Giles
- School of Pharmacy and Medical Sciences; University of South Australia; Adelaide Australia
| | - T. Vanniasinkam
- School of Biomedical Sciences; Charles Sturt University; Wagga Wagga New South Wales Australia
| | - S. Ndi
- School of Pharmacy and Medical Sciences; University of South Australia; Adelaide Australia
| | - M. D. Barton
- School of Pharmacy and Medical Sciences; University of South Australia; Adelaide Australia
| |
Collapse
|
26
|
de Cassan SC, Draper SJ. Recent advances in antibody-inducing poxviral and adenoviral vectored vaccine delivery platforms for difficult disease targets. Expert Rev Vaccines 2014; 12:365-78. [DOI: 10.1586/erv.13.11] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
27
|
Induction of antigen-positive cell death by the expression of perforin, but not DTa, from a DNA vaccine enhances the immune response. Immunol Cell Biol 2013; 92:359-67. [PMID: 24323081 DOI: 10.1038/icb.2013.93] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 10/28/2013] [Accepted: 11/09/2013] [Indexed: 02/06/2023]
Abstract
The failure of traditional protein-based vaccines to prevent infection by viruses such as HIV or hepatitis C highlights the need for novel vaccine strategies. DNA vaccines have shown promise in small animal models, and are effective at generating anti-viral T cell-mediated immune responses; however, they have proved to be poorly immunogenic in clinical trials. We propose that the induction of necrosis will enhance the immune response to vaccine antigens encoded by DNA vaccines, as necrotic cells are known to release a range of intracellular factors that lead to dendritic cell (DC) activation and enhanced cross-presentation of antigen. Here we provide evidence that induction of cell death in DNA vaccine-targeted cells provides an adjuvant effect following intradermal vaccination of mice; however, this enhancement of the immune response is dependent on both the mechanism and timing of cell death after antigen expression. We report that a DNA vaccine encoding the cytolytic protein, perforin, resulted in DC activation, enhanced broad and multifunctional CD8 T-cell responses to the HIV-1 antigen GAG and reduced viral load following challenge with a chimeric virus, EcoHIV, compared with the canonical GAG DNA vaccine. This effect was not observed for a DNA vaccine encoding an apoptosis-inducing toxin, DTa, or when the level of perforin expression was increased to induce cell death sooner after vaccination. Thus, inducing lytic cell death following a threshold level of expression of a viral antigen can improve the immunogenicity of DNA vaccines, whereas apoptotic cell death has an inhibitory effect on the immune response.
Collapse
|
28
|
Immunological characterization of a modified vaccinia virus Ankara vector expressing the human papillomavirus 16 E1 protein. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2013; 21:147-55. [PMID: 24307238 DOI: 10.1128/cvi.00678-13] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Women showing normal cytology but diagnosed with a persistent high-risk human papillomavirus (HR-HPV) infection have a higher risk of developing high-grade cervical intraepithelial neoplasia and cervical cancer than noninfected women. As no therapeutic management other than surveillance is offered to these women, there is a major challenge to develop novel targeted therapies dedicated to the treatment of these patients. As such, E1 and E2 antigens, expressed early in the HPV life cycle, represent very interesting candidates. Both proteins are necessary for maintaining coordinated viral replication and gene synthesis during the differentiation process of the epithelium and are essential for the virus to complete its normal and propagative replication cycle. In the present study, we evaluated a new active targeted immunotherapeutic, a modified vaccinia virus Ankara (MVA) vector containing the E1 sequence of HPV16, aimed at inducing cellular immune responses with the potential to help and clear persistent HPV16-related infection. We carried out an extensive comparative time course analysis of the cellular immune responses induced by different schedules of immunization in C57BL/6 mice. We showed that multiple injections of MVA-E1 allowed sustained HPV16 E1-specific cellular immune responses in vaccinated mice and had no impact on the exhaustion phenotype of the generated HPV16 E1-specific CD8⁺ T cells, but they led to the differentiation of multifunctional effector T cells with high cytotoxic capacity. This study provides proof of concept that an MVA expressing HPV16 E1 can induce robust and long-lasting E1-specific responses and warrants further development of this candidate.
Collapse
|
29
|
Draper SJ, Cottingham MG, Gilbert SC. Utilizing poxviral vectored vaccines for antibody induction-progress and prospects. Vaccine 2013; 31:4223-30. [PMID: 23746455 PMCID: PMC7131268 DOI: 10.1016/j.vaccine.2013.05.091] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 05/22/2013] [Indexed: 02/06/2023]
Abstract
Poxviral vectors are now regarded as robust tools for B cell and antibody induction. Antibody responses can be induced against the vector as well as a transgene. Increasing application is seen in heterologous prime–boost immunization regimes. Effective veterinary poxviral vaccine products are now licensed. Promising results of antibody induction are being reported in human clinical trials.
Over the last decade, poxviral vectors emerged as a mainstay approach for the induction of T cell-mediated immunity by vaccination, and their suitability for human use has led to widespread clinical testing of candidate vectors against infectious intracellular pathogens and cancer. In contrast, poxviruses have been widely perceived in the vaccine field as a poor choice of vector for the induction of humoral immunity. However, a growing body of data, from both animal models and recent clinical trials, now suggests that these vectors can be successfully utilized to prime and boost B cells and effective antibody responses. Significant progress has been made in the context of heterologous prime–boost immunization regimes, whereby poxviruses are able to boost responses primed by other vectors, leading to the induction of high-titre antigen-specific antibody responses. In other cases, poxviral vectors have been shown to stimulate humoral immunity against both themselves and encoded transgenes, in particular viral surface proteins such as influenza haemagglutinin. In the veterinary field, recombinant poxviral vectors have made a significant impact with numerous vectors licensed for use against a variety of animal viruses. On-going studies continue to explore the potential of poxviral vectors to modulate qualitative aspects of the humoral response, as well as their amenability to adjuvantation seeking to improve quantitative antibody immunogenicity. Nevertheless, the underlying mechanisms of B cell induction by recombinant poxviruses remain poorly defined, and further work is necessary to help guide the rational optimization of future poxviral vaccine candidates aiming to induce antibodies.
Collapse
Affiliation(s)
- Simon J Draper
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK.
| | | | | |
Collapse
|
30
|
Starodubova E, Krotova O, Hallengärd D, Kuzmenko Y, Engström G, Legzdina D, Latyshev O, Eliseeva O, Maltais AK, Tunitskaya V, Karpov V, Bråve A, Isaguliants M. Cellular Immunogenicity of Novel Gene Immunogens in Mice Monitored by in Vivo Imaging. Mol Imaging 2012. [DOI: 10.2310/7290.2012.00011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Elizaveta Starodubova
- From the Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden; WA Engelhardt Institute of Molecular Biology, Moscow, Russia; Center of Medical Research, University of Oslo, Moscow, Russia; DI Ivanovsky Institute of Virology, Moscow, Russia; and Cytopulse AB, Stockholm, Sweden
| | - Olga Krotova
- From the Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden; WA Engelhardt Institute of Molecular Biology, Moscow, Russia; Center of Medical Research, University of Oslo, Moscow, Russia; DI Ivanovsky Institute of Virology, Moscow, Russia; and Cytopulse AB, Stockholm, Sweden
| | - David Hallengärd
- From the Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden; WA Engelhardt Institute of Molecular Biology, Moscow, Russia; Center of Medical Research, University of Oslo, Moscow, Russia; DI Ivanovsky Institute of Virology, Moscow, Russia; and Cytopulse AB, Stockholm, Sweden
| | - Yulia Kuzmenko
- From the Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden; WA Engelhardt Institute of Molecular Biology, Moscow, Russia; Center of Medical Research, University of Oslo, Moscow, Russia; DI Ivanovsky Institute of Virology, Moscow, Russia; and Cytopulse AB, Stockholm, Sweden
| | - Gunnel Engström
- From the Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden; WA Engelhardt Institute of Molecular Biology, Moscow, Russia; Center of Medical Research, University of Oslo, Moscow, Russia; DI Ivanovsky Institute of Virology, Moscow, Russia; and Cytopulse AB, Stockholm, Sweden
| | - Diana Legzdina
- From the Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden; WA Engelhardt Institute of Molecular Biology, Moscow, Russia; Center of Medical Research, University of Oslo, Moscow, Russia; DI Ivanovsky Institute of Virology, Moscow, Russia; and Cytopulse AB, Stockholm, Sweden
| | - Oleg Latyshev
- From the Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden; WA Engelhardt Institute of Molecular Biology, Moscow, Russia; Center of Medical Research, University of Oslo, Moscow, Russia; DI Ivanovsky Institute of Virology, Moscow, Russia; and Cytopulse AB, Stockholm, Sweden
| | - Olesja Eliseeva
- From the Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden; WA Engelhardt Institute of Molecular Biology, Moscow, Russia; Center of Medical Research, University of Oslo, Moscow, Russia; DI Ivanovsky Institute of Virology, Moscow, Russia; and Cytopulse AB, Stockholm, Sweden
| | - Anna Karin Maltais
- From the Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden; WA Engelhardt Institute of Molecular Biology, Moscow, Russia; Center of Medical Research, University of Oslo, Moscow, Russia; DI Ivanovsky Institute of Virology, Moscow, Russia; and Cytopulse AB, Stockholm, Sweden
| | - Vera Tunitskaya
- From the Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden; WA Engelhardt Institute of Molecular Biology, Moscow, Russia; Center of Medical Research, University of Oslo, Moscow, Russia; DI Ivanovsky Institute of Virology, Moscow, Russia; and Cytopulse AB, Stockholm, Sweden
| | - Vadim Karpov
- From the Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden; WA Engelhardt Institute of Molecular Biology, Moscow, Russia; Center of Medical Research, University of Oslo, Moscow, Russia; DI Ivanovsky Institute of Virology, Moscow, Russia; and Cytopulse AB, Stockholm, Sweden
| | - Andreas Bråve
- From the Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden; WA Engelhardt Institute of Molecular Biology, Moscow, Russia; Center of Medical Research, University of Oslo, Moscow, Russia; DI Ivanovsky Institute of Virology, Moscow, Russia; and Cytopulse AB, Stockholm, Sweden
| | - Maria Isaguliants
- From the Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden; WA Engelhardt Institute of Molecular Biology, Moscow, Russia; Center of Medical Research, University of Oslo, Moscow, Russia; DI Ivanovsky Institute of Virology, Moscow, Russia; and Cytopulse AB, Stockholm, Sweden
| |
Collapse
|
31
|
Lambe T. Novel viral vectored vaccines for the prevention of influenza. Mol Med 2012; 18:1153-60. [PMID: 22735755 PMCID: PMC3510293 DOI: 10.2119/molmed.2012.00147] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Accepted: 06/19/2012] [Indexed: 01/29/2023] Open
Abstract
Influenza represents a substantial global healthcare burden, with annual epidemics resulting in 3-5 million cases of severe illness with a significant associated mortality. In addition, the risk of a virulent and lethal influenza pandemic has generated widespread and warranted concern. Currently licensed influenza vaccines are limited in their ability to induce efficacious and long-lasting herd immunity. In addition, and as evidenced by the H1N1 pandemic in 2009, there can be a significant delay between the emergence of a pandemic influenza and an effective, antibody-inducing vaccine. There is, therefore, a continued need for new, efficacious vaccines conferring cross-clade protection-obviating the need for biannual reformulation of seasonal influenza vaccines. Development of such a vaccine would yield enormous health benefits to society and also greatly reduce the associated global healthcare burden. There are a number of alternative influenza vaccine technologies being assessed both preclinically and clinically. In this review we discuss viral vectored vaccines, either recombinant live-attenuated or replication-deficient viruses, which are current lead candidates for inducing efficacious and long-lasting immunity toward influenza viruses. These alternate influenza vaccines offer real promise to deliver viable alternatives to currently deployed vaccines and more importantly may confer long-lasting and universal protection against influenza viral infection.
Collapse
Affiliation(s)
- Teresa Lambe
- Jenner Institute, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
32
|
You Q, Wu Y, Wu Y, Wei W, Wang C, Jiang D, Yu X, Zhang X, Wang Y, Tang Z, Jiang C, Kong W. Immunogenicity and protective efficacy of heterologous prime-boost regimens with mycobacterial vaccines and recombinant adenovirus- and poxvirus-vectored vaccines against murine tuberculosis. Int J Infect Dis 2012; 16:e816-25. [PMID: 22921259 DOI: 10.1016/j.ijid.2012.07.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 07/03/2012] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVES To evaluate regimens using bacillus Calmette-Guérin (BCG) or recombinant BCG (rBCG) overexpressing Ag85B for priming, followed by boosting with a modified vaccinia virus Ankara strain (MVA) and/or adenovirus vector (AD) expressing an Ag85B-ESAT6 fusion protein. METHODS Cellular and humoral immune responses were determined after subcutaneous vaccination, which was employed to trigger systemic immunity against intravenous infection in a mouse model of tuberculosis (TB). Bacterial loads and lung histology were evaluated. RESULTS The relative IgG2a and IgG1 antibody levels indicated that the viral-vectored vaccines generated a T-helper type 1 (Th1)-biased response after two doses of viral boost vaccinations. Boosting BCG-primed mice with viral vaccines induced a Th1 immune response that included both CD4 and CD8 T-cells generating antigen-specific interferon-gamma (IFN-γ) and CD8 T cytotoxic activity. Only mice vaccinated with two different viral boosters after BCG priming exhibited a significant reduction in bacterial burden in the lung after challenge. Histology examinations confirmed the attenuation of lung damage and more compact granulomas. After mycobacteria priming, boosting with AD85B-E6 followed by MVA85B-E6 afforded better protection than the reverse order of administration of the viral vectors. CONCLUSIONS This study demonstrates the potential of multiple heterologous viral booster vaccines, although the exact correlates of protection and optimal regimens should be further investigated for the rational design of future vaccine strategies.
Collapse
Affiliation(s)
- Qingrui You
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Gaoxin District Changchun, Jilin, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Guzman E, Cubillos-Zapata C, Cottingham MG, Gilbert SC, Prentice H, Charleston B, Hope JC. Modified vaccinia virus Ankara-based vaccine vectors induce apoptosis in dendritic cells draining from the skin via both the extrinsic and intrinsic caspase pathways, preventing efficient antigen presentation. J Virol 2012; 86:5452-66. [PMID: 22419811 PMCID: PMC3347273 DOI: 10.1128/jvi.00264-12] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 03/05/2012] [Indexed: 02/03/2023] Open
Abstract
Dendritic cells (DC) are potent antigen-presenting cells and central to the induction of immune responses following infection or vaccination. The collection of DC migrating from peripheral tissues by cannulation of the afferent lymphatic vessels provides DC which can be used directly ex vivo without extensive in vitro manipulations. We have previously used bovine migrating DC to show that recombinant human adenovirus 5 vectors efficiently transduce afferent lymph migrating DEC-205(+) CD11c(+) CD8(-) DC (ALDC). We have also shown that recombinant modified vaccinia virus Ankara (MVA) infects ALDC in vitro, causing downregulation of costimulatory molecules, apoptosis, and cell death. We now show that in the bovine system, modified vaccinia virus Ankara-induced apoptosis in DC draining from the skin occurs soon after virus binding via the caspase 8 pathway and is not associated with viral gene expression. We also show that after virus entry, the caspase 9 pathway cascade is initiated. The magnitude of T cell responses to mycobacterial antigen 85A (Ag85A) expressed by recombinant MVA-infected ALDC is increased by blocking caspase-induced apoptosis. Apoptotic bodies generated by recombinant MVA (rMVA)-Ag85A-infected ALDC and containing Ag85A were phagocytosed by noninfected migrating ALDC expressing SIRPα via actin-dependent phagocytosis, and these ALDC in turn presented antigen. However, the addition of fresh ALDC to MVA-infected cultures did not improve on the magnitude of the T cell responses; in contrast, these noninfected DC showed downregulation of major histocompatibility complex class II (MHC-II), CD40, CD80, and CD86. We also observed that MVA-infected ALDC promoted migration of DEC-205(+) SIRPα(+) CD21(+) DC as well as CD4(+) and CD8(+) T cells independently of caspase activation. These in vitro studies show that induction of apoptosis in DC by MVA vectors is detrimental to the subsequent induction of T cell responses.
Collapse
Affiliation(s)
- E Guzman
- Institute for Animal Health, Compton, Newbury, Berkshire, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
34
|
Jackson SS, Schmitz JE, Letvin NL. Anti-gamma interferon antibodies enhance the immunogenicity of recombinant adenovirus vectors. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2011; 18:1969-78. [PMID: 21900534 PMCID: PMC3209036 DOI: 10.1128/cvi.05180-11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Accepted: 08/29/2011] [Indexed: 11/20/2022]
Abstract
Vaccination for eliciting antigen-specific memory CD8(+) T cells may be facilitated by manipulating the pleiotropic effects of gamma interferon (IFN-γ). We assessed strategies for modulating the contribution of IFN-γ during the development of antigen-specific cytotoxic T lymphocyte (CTL) populations. We first showed that recombinant IFN-γ suppressed antigen expression in vitro from a recombinant adenovirus (rAd) vector in a dose-dependent manner and that addition of an anti-IFN-γ antibody (Ab) eliminated this suppression. Consistent with these in vitro findings, we found that HIV-1 envelope (Env)-specific CTL responses were higher in IFN-γ-knockout (GKO) mice than in wild-type mice following immunization with rAd. Since these observations suggested that IFN-γ might suppress rAd-induced CTL development, we assessed the ability of anti-IFN-γ Ab administration to augment rAd-elicited CTL in vivo. In fact, blockage of IFN-γ activity by monoclonal Ab administration was associated with elevated levels of interleukin 7 receptor alpha chain-positive (IL-7Rα(+)) Env-specific CTL populations postboost. These observations illustrate the utility of an anti-IFN-γ Ab for potentiating rAd immunizations to effect quantitative and qualitative changes in the effector and memory CTL populations.
Collapse
MESH Headings
- AIDS Vaccines/administration & dosage
- AIDS Vaccines/genetics
- AIDS Vaccines/immunology
- Adenoviridae/genetics
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/genetics
- Animals
- Antibodies/administration & dosage
- Antibodies/immunology
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/genetics
- CD8-Positive T-Lymphocytes/immunology
- Female
- Genetic Vectors
- HIV-1/genetics
- HIV-1/immunology
- Interferon-gamma/antagonists & inhibitors
- Mice
- Mice, Inbred BALB C
- T-Lymphocytes, Cytotoxic/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- env Gene Products, Human Immunodeficiency Virus/genetics
- env Gene Products, Human Immunodeficiency Virus/immunology
Collapse
Affiliation(s)
| | - Jörn E. Schmitz
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Norman L. Letvin
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
35
|
Sedegah M, Tamminga C, McGrath S, House B, Ganeshan H, Lejano J, Abot E, Banania GJ, Sayo R, Farooq F, Belmonte M, Manohar N, Richie NO, Wood C, Long CA, Regis D, Williams FT, Shi M, Chuang I, Spring M, Epstein JE, Mendoza-Silveiras J, Limbach K, Patterson NB, Bruder JT, Doolan DL, King CR, Soisson L, Diggs C, Carucci D, Dutta S, Hollingdale MR, Ockenhouse CF, Richie TL. Adenovirus 5-vectored P. falciparum vaccine expressing CSP and AMA1. Part A: safety and immunogenicity in seronegative adults. PLoS One 2011; 6:e24586. [PMID: 22003383 PMCID: PMC3189181 DOI: 10.1371/journal.pone.0024586] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Accepted: 08/15/2011] [Indexed: 11/24/2022] Open
Abstract
Background Models of immunity to malaria indicate the importance of CD8+ T cell responses for targeting intrahepatic stages and antibodies for targeting sporozoite and blood stages. We designed a multistage adenovirus 5 (Ad5)-vectored Plasmodium falciparum malaria vaccine, aiming to induce both types of responses in humans, that was tested for safety and immunogenicity in a Phase 1 dose escalation trial in Ad5-seronegative volunteers. Methodology/Principal Findings The NMRC-M3V-Ad-PfCA vaccine combines two adenovectors encoding circumsporozoite protein (CSP) and apical membrane antigen-1 (AMA1). Group 1 (n = 6) healthy volunteers received one intramuscular injection of 2×10∧10 particle units (1×10∧10 each construct) and Group 2 (n = 6) a five-fold higher dose. Transient, mild to moderate adverse events were more pronounced with the higher dose. ELISpot responses to CSP and AMA1 peaked at 1 month, were higher in the low dose (geomean CSP = 422, AMA1 = 862 spot forming cells/million) than in the high dose (CSP = 154, p = 0.049, AMA1 = 423, p = 0.045) group and were still positive at 12 months in a number of volunteers. ELISpot depletion assays identified dependence on CD4+ or on both CD4+ and CD8+ T cells, with few responses dependent only on CD8+ T cells. Intracellular cytokine staining detected stronger CD8+ than CD4+ T cell IFN-γ responses (CSP p = 0.0001, AMA1 p = 0.003), but similar frequencies of multifunctional CD4+ and CD8+ T cells secreting two or more of IFN-γ, TNF-α or IL-2. Median fluorescence intensities were 7–10 fold higher in triple than single secreting cells. Antibody responses were low but trended higher in the high dose group and did not inhibit growth of cultured P. falciparum blood stage parasites. Significance As found in other trials, adenovectored vaccines appeared safe and well-tolerated at doses up to 1×10∧11 particle units. This is the first demonstration in humans of a malaria vaccine eliciting strong CD8+ T cell IFN-γ responses. Trial Registration ClinicalTrials.govNCT00392015
Collapse
Affiliation(s)
- Martha Sedegah
- U.S. Military Malaria Vaccine Program, Naval Medical Research Center, Silver Spring, Maryland, United States of America.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
de Cassan SC, Forbes EK, Douglas AD, Milicic A, Singh B, Gupta P, Chauhan VS, Chitnis CE, Gilbert SC, Hill AVS, Draper SJ. The requirement for potent adjuvants to enhance the immunogenicity and protective efficacy of protein vaccines can be overcome by prior immunization with a recombinant adenovirus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 187:2602-16. [PMID: 21813775 PMCID: PMC3160495 DOI: 10.4049/jimmunol.1101004] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
A central goal in vaccinology is the induction of high and sustained Ab responses. Protein-in-adjuvant formulations are commonly used to achieve such responses. However, their clinical development can be limited by the reactogenicity of some of the most potent preclinical adjuvants and the cost and complexity of licensing new adjuvants for human use. Also, few adjuvants induce strong cellular immunity, which is important for protection against many diseases, such as malaria. We compared classical adjuvants such as aluminum hydroxide to new preclinical adjuvants and adjuvants in clinical development, such as Abisco 100, CoVaccine HT, Montanide ISA720, and stable emulsion-glucopyranosyl lipid A, for their ability to induce high and sustained Ab responses and T cell responses. These adjuvants induced a broad range of Ab responses when used in a three-shot protein-in-adjuvant regimen using the model Ag OVA and leading blood-stage malaria vaccine candidate Ags. Surprisingly, this range of Ab immunogenicity was greatly reduced when a protein-in-adjuvant vaccine was used to boost Ab responses primed by a human adenovirus serotype 5 vaccine recombinant for the same Ag. This human adenovirus serotype 5-protein regimen also induced a more cytophilic Ab response and demonstrated improved efficacy of merozoite surface protein-1 protein vaccines against a Plasmodium yoelii blood-stage challenge. This indicates that the differential immunogenicity of protein vaccine adjuvants may be largely overcome by prior immunization with recombinant adenovirus, especially for adjuvants that are traditionally considered poorly immunogenic in the context of subunit vaccination and may circumvent the need for more potent chemical adjuvants.
Collapse
Affiliation(s)
- Simone C de Cassan
- The Jenner Institute, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Reyes-Sandoval A, Wyllie DH, Bauza K, Milicic A, Forbes EK, Rollier CS, Hill AVS. CD8+ T effector memory cells protect against liver-stage malaria. THE JOURNAL OF IMMUNOLOGY 2011; 187:1347-57. [PMID: 21715686 DOI: 10.4049/jimmunol.1100302] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Identification of correlates of protection for infectious diseases including malaria is a major challenge and has become one of the main obstacles in developing effective vaccines. We investigated protection against liver-stage malaria conferred by vaccination with adenoviral (Ad) and modified vaccinia Ankara (MVA) vectors expressing pre-erythrocytic malaria Ags. By classifying CD8(+) T cells into effector, effector memory (T(EM)), and central memory subsets using CD62L and CD127 markers, we found striking differences in T cell memory generation. Although MVA induced accelerated central memory T cell generation, which could be efficiently boosted by subsequent Ad administration, it failed to protect against malaria. In contrast, Ad vectors, which permit persistent Ag delivery, elicit a prolonged effector T cell and T(EM) response that requires long intervals for an efficient boost. A preferential T(EM) phenotype was maintained in liver, blood, and spleen after Ad/MVA prime-boost regimens, and animals were protected against malaria sporozoite challenge. Blood CD8(+) T(EM) cells correlated with protection against malaria liver-stage infection, assessed by estimation of number of parasites emerging from the liver into the blood. The protective ability of Ag-specific T(EM) cells was confirmed by transfer experiments into naive recipient mice. Thus, we identify persistent CD8 T(EM) populations as essential for vaccine-induced pre-erythrocytic protection against malaria, a finding that has important implications for vaccine design.
Collapse
|
38
|
Potent and broadly reactive HIV-2 neutralizing antibodies elicited by a vaccinia virus vector prime-C2V3C3 polypeptide boost immunization strategy. J Virol 2010; 84:12429-36. [PMID: 20844029 DOI: 10.1128/jvi.01102-10] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Human immunodeficiency virus type 2 (HIV-2) infection affects about 1 to 2 million individuals, the majority living in West Africa, Europe, and India. As for HIV-1, new strategies for the prevention of HIV-2 infection are needed. Our aim was to produce new vaccine immunogens that elicit the production of broadly reactive HIV-2 neutralizing antibodies (NAbs). Native and truncated envelope proteins from the reference HIV-2ALI isolate were expressed in vaccinia virus or in bacteria. This source isolate was used due to its unique phenotype combining CD4 independence and CCR5 usage. NAbs were not elicited in BALB/c mice by single immunization with a truncated and fully glycosylated envelope gp125 (gp125t) or a recombinant polypeptide comprising the C2, V3, and C3 envelope regions (rpC2-C3). A strong and broad NAb response was, however, elicited in mice primed with gp125t expressed in vaccinia virus and boosted with rpC2-C3. Serum from these animals potently neutralized (median 50% neutralizing titer, 3,200) six of six highly divergent primary HIV-2 isolates. Coreceptor usage and the V3 sequence of NAb-sensitive isolates were similar to that of the vaccinating immunogen (HIV-2ALI). In contrast, NAbs were not reactive on three X4 isolates that displayed major changes in V3 loop sequence and structure. Collectively, our findings demonstrate that broadly reactive HIV-2 NAbs can be elicited by using a vaccinia virus vector-prime/rpC2-C3-boost immunization strategy and suggest a potential relationship between escape to neutralization and cell tropism.
Collapse
|
39
|
Graham BS, Kines R, Corbett KS, Nicewonger J, Johnson TR, Chen M, LaVigne D, Roberts JN, Cuburu N, Schiller JT, Buck CB. Mucosal delivery of human papillomavirus pseudovirus-encapsidated plasmids improves the potency of DNA vaccination. Mucosal Immunol 2010; 3:475-86. [PMID: 20555315 PMCID: PMC2924464 DOI: 10.1038/mi.2010.31] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Mucosal immunization may be important for protection against pathogens whose transmission and pathogenesis target the mucosal tissue. The capsid proteins of human papillomavirus (HPV) confer tropism for the basal epithelium and can encapsidate DNA during self-assembly to form pseudovirions (PsVs). Therefore, we produced mucosal vaccine vectors by HPV PsV encapsidation of DNA plasmids expressing an experimental antigen derived from the M and M2 proteins of respiratory syncytial virus (RSV). Intravaginal (IVag) delivery elicited local and systemic M-M2-specific CD8+ T-cell and antibody responses in mice that were comparable to an approximately 10,000-fold higher dose of naked DNA. A single HPV PsV IVag immunization primed for M-M2-specific-IgA in nasal and vaginal secretions. Based on light emission and immunofluorescent microscopy, immunization with HPV PsV-encapsidated luciferase- and red fluorescent protein (RFP)-expressing plasmids resulted in transient antigen expression (<5 days), which was restricted to the vaginal epithelium. HPV PsV encapsidation of plasmid DNA is a novel strategy for mucosal immunization that could provide new vaccine options for selected mucosal pathogens.
Collapse
Affiliation(s)
- Barney S. Graham
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Rhonda Kines
- Laboratory of Cellular Oncology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Kizzmekia S. Corbett
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - John Nicewonger
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Teresa R. Johnson
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Man Chen
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Daaimah LaVigne
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | | | - Nicolas Cuburu
- Laboratory of Cellular Oncology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - John T. Schiller
- Laboratory of Cellular Oncology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Christopher B. Buck
- Laboratory of Cellular Oncology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
40
|
Induction of both cellular and humoral immunity following a rational prime-boost immunization regimen that incorporates recombinant ovine atadenovirus and fowlpox virus. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2010; 17:1679-86. [PMID: 20810681 DOI: 10.1128/cvi.00291-10] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Recombinant fowlpox viruses (rFPV) and ovine atadenoviruses (rOAdV) are being developed as safe, nonpathogenic, prophylactic and therapeutic vaccine vectors. There is scope, however, to improve the limited immune responses elicited by each of these vaccine vectors. Using previously determined and optimized routes of administration and viral doses, we characterized the primary adaptive immune responses elicited by recombinant variants of each virus. We demonstrate the contrasting nature of the response elicited by each recombinant virus. Whereas rFPV generates predominately cell-mediated immunity to our nominal target antigen, ovalbumin (OVA), rOAdV drives strong humoral responses. By defining the time taken to achieve maximal cytotoxic T cell responses and by studying the different patterns and kinetics of major histocompatibility complex class I-restricted OVA antigen expression postimmunization, we proposed a heterologous prime-boost regimen of immunization with rOAdV followed by rFPV. The subsequent experimental results showed that this approach produced robust cell-mediated and humoral immune responses against OVA that, importantly, were accompanied by weak anti-viral vector antibody responses. These results, therefore, represent a novel and potentially clinically applicable way to achieve broadly based and effective immunity to the antigens encoded by vectored vaccines.
Collapse
|
41
|
Frimpong-Boateng K, van Rooijen N, Geiben-Lynn R. Regulatory T cells suppress natural killer cells during plasmid DNA vaccination in mice, blunting the CD8+ T cell immune response by the cytokine TGFbeta. PLoS One 2010; 5:e12281. [PMID: 20808850 PMCID: PMC2924372 DOI: 10.1371/journal.pone.0012281] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2010] [Accepted: 07/23/2010] [Indexed: 01/21/2023] Open
Abstract
Background CD4+CD25+ regulatory T cells (Tregs) suppress adaptive T cell-mediated immune responses to self- and foreign-antigens. Tregs may also suppress early innate immune responses to vaccine antigens and might decrease vaccine efficacy. NK and NKT cells are the first responders after plasmid DNA vaccination and are found at the site of inoculation. Earlier reports demonstrated that NKT cells could improve plasmid DNA efficacy, a phenomenon not found for NK cells. In fact, it has been shown that under certain disease conditions, NK cells are suppressed by Tregs via their release of IL-10 and/or TGFβ. Therefore, we tested the hypothesis that NK cell function is suppressed by Tregs in the setting of plasmid DNA vaccination. Methodology/Principal Findings In this study we show that Tregs directly inhibit NK cell function during plasmid DNA vaccination by suppressing the potentially 10-fold, NK cell-mediated, augmentation of plasmid DNA antigen-specific CD8+ T cells. We found that this phenomenon is dependent on the secretion of cytokine TGFβ by Tregs, and independent of IL-10. Conclusions Our data indicate a crucial function for Tregs in blocking plasmid DNA vaccine-elicited immune responses, revealing potentially novel strategies for improving the efficiency of plasmid DNA vaccines including chemical- or antibody-induced localized blockage of Treg-mediated suppression of NK cells at the site of plasmid DNA vaccine inoculation.
Collapse
Affiliation(s)
- Kwesi Frimpong-Boateng
- Division of Viral Pathogenesis, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Nico van Rooijen
- Department of Molecular Cell Biology, Vrije University Medical Center, Amsterdam, The Netherlands
| | - Ralf Geiben-Lynn
- Division of Viral Pathogenesis, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
42
|
Draper SJ, Heeney JL. Viruses as vaccine vectors for infectious diseases and cancer. Nat Rev Microbiol 2010; 8:62-73. [PMID: 19966816 DOI: 10.1038/nrmicro2240] [Citation(s) in RCA: 261] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Recent developments in the use of viruses as vaccine vectors have been facilitated by a better understanding of viral biology. Advances occur as we gain greater insight into the interrelationship of viruses and the immune system. Viral-vector vaccines remain the best means to induce cellular immunity and are now showing promise for the induction of strong humoral responses. The potential benefits for global health that are offered by this field reflect the scope and utility of viruses as vaccine vectors for human and veterinary applications, with targets ranging from certain types of cancer to a vast array of infectious diseases.
Collapse
Affiliation(s)
- Simon J Draper
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK.
| | | |
Collapse
|
43
|
Geiben-Lynn R, Greenland JR, Frimpong-Boateng K, Letvin NL. Non-classical natural killer T cells modulate plasmid DNA vaccine antigen expression and vaccine-elicited immune responses by MCP-1 secretion after interaction with a beta2-microglobulin-independent CD1d. J Biol Chem 2009; 284:33800-6. [PMID: 19833737 DOI: 10.1074/jbc.m109.019638] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The magnitude and durability of a plasmid DNA vaccine-induced immune response is shaped by immune effector molecules at the site of vaccination. In the present study, we show that antigen expression is modified by type II NKT cells, after interaction with a beta2-microglobulin-independent CD1d receptor. After activation, during the first days following plasmid DNA vaccination, NKT cells release IL-5 and MCP-1, leading to a T helper 0 (T(H)0) cytokine/chemokine profile and a stronger CD8(+)/CD4(+) T cell immune response. Our data indicate that this phenomenon was induced through the strong T(H)1 chemokine MCP-1 during the early phases of plasmid DNA vaccination because injecting the type II NKT cell-associated MCP-1 during the first 5 days led to 2-3-fold increases in vaccine-elicited T cell responses. This study demonstrates a critical role for NKT cells in plasmid DNA vaccine-induced immune responses. Manipulation of NKT cell function or co-administration of MCP-1 may represent novel methods for enhancing immune responses to plasmid DNA vaccines.
Collapse
Affiliation(s)
- Ralf Geiben-Lynn
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, and Harvard Medical School, Boston, Massachusetts 02215, USA.
| | | | | | | |
Collapse
|