1
|
Rzymski P, Szuster-Ciesielska A, Dzieciątkowski T, Gwenzi W, Fal A. mRNA vaccines: The future of prevention of viral infections? J Med Virol 2023; 95:e28572. [PMID: 36762592 DOI: 10.1002/jmv.28572] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023]
Abstract
Messenger RNA (mRNA) vaccines against COVID-19 are the first authorized biological preparations developed using this platform. During the pandemic, their administration has been proven to be a life-saving intervention. Here, we review the main advantages of using mRNA vaccines, identify further technological challenges to be met during the development of the mRNA platform, and provide an update on the clinical progress on leading mRNA vaccine candidates against different viruses that include influenza viruses, human immunodeficiency virus 1, respiratory syncytial virus, Nipah virus, Zika virus, human cytomegalovirus, and Epstein-Barr virus. The prospects and challenges of manufacturing mRNA vaccines in low-income countries are also discussed. The ongoing interest and research in mRNA technology are likely to overcome some existing challenges for this technology (e.g., related to storage conditions and immunogenicity of some components of lipid nanoparticles) and enhance the portfolio of vaccines against diseases for which classical formulations are already authorized. It may also open novel pathways of protection against infections and their consequences for which no safe and efficient immunization methods are currently available.
Collapse
Affiliation(s)
- Piotr Rzymski
- Department of Environmental Medicine, Poznan University of Medical Sciences, Poznań, Poland.,Integrated Science Association (ISA), Universal Scientific Education and Research Network (USERN), Poznań, Poland
| | - Agnieszka Szuster-Ciesielska
- Department of Virology and Immunology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Lublin, Poland
| | | | - Willis Gwenzi
- Alexander von Humboldt Fellow & Guest Professor, Grassland Science and Renewable Plant Resources, Faculty of Organic Agricultural Sciences, Universität Kassel, Witzenhausen, Germany.,Alexander von Humboldt Fellow & Guest Professor, Leibniz Institute for Agricultural Engineering and Bioeconomy (ATB), Potsdam, Germany
| | - Andrzej Fal
- Collegium Medicum, Warsaw Faculty of Medicine, Cardinal Stefan Wyszynski University, Warsaw, Poland.,Department of Public Health, Wrocław Medical University, Wrocław, Poland
| |
Collapse
|
2
|
Tarimo EAM, Ambikile J, Munseri P, Bakari M. Personal experiences following acquiring HIV infection while volunteering in Phase I/II HIV vaccine trials: A qualitative study from Tanzania. PLoS One 2022; 17:e0276404. [PMID: 36288332 PMCID: PMC9605023 DOI: 10.1371/journal.pone.0276404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 10/05/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Participation in HIV vaccine trials is an essential step towards development of an effective preventive vaccine. A Phase I/II HIV vaccine trial enrolls volunteers at low risk of acquiring HIV infection, however a few may still become infected. Understanding the experiences of volunteers who acquired HIV infection while participating in such trials is essential for future research. Here, we describe experiences of HIV infected volunteers in Phase I/II HIV vaccine trials conducted in urban Tanzania. MATERIALS AND METHODS We used a case study design. In-depth interviews were conducted with four participants who became HIV infected during long follow-up visits after completion of vaccination schedules in a Phase I/II trial. Between 3 and 8 years after HIV positive diagnosis, each participant was interviewed at three time points within a two-year interval so as to allow for accumulation of experiences and cross-checking the emerging constructs. Data was analyzed using a qualitative data analysis framework. RESULTS Analysis revealed that participation in HIV vaccine trials involves balancing controversies and the spirit of informed decision. The participants declared that they did not acquire HIV from the experimental vaccine. Disclosure of HIV status within the family was gender specific. Men were hesitant to disclose their HIV status to their sexual partners fearing for the consequences. Women's attempt to disclose their HIV status yielded negative reactions from the sexual partners. The acquired knowledge from the HIV vaccine research enabled the participants to cope with the uncertainties and their health status. CONCLUSIONS The knowledge acquired during the Phase I/II HIV vaccine trial appears to be an essential resource to cope with uncertainties post research. The HIV vaccine trial implementers need to understand the challenges the volunteers may confront after the trial while coping with their health status. Longitudinal studies are essential to trace the effects of uncertainties to the individual participants.
Collapse
Affiliation(s)
- Edith A. M. Tarimo
- Department of Nursing Management, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Joel Ambikile
- Department of Clinical Nursing, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Patricia Munseri
- Department of Internal Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Muhammad Bakari
- Department of Internal Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| |
Collapse
|
3
|
Ng'uni T, Chasara C, Ndhlovu ZM. Major Scientific Hurdles in HIV Vaccine Development: Historical Perspective and Future Directions. Front Immunol 2020; 11:590780. [PMID: 33193428 PMCID: PMC7655734 DOI: 10.3389/fimmu.2020.590780] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/05/2020] [Indexed: 12/15/2022] Open
Abstract
Following the discovery of HIV as a causative agent of AIDS, the expectation was to rapidly develop a vaccine; but thirty years later, we still do not have a licensed vaccine. Progress has been hindered by the extensive genetic variability of HIV and our limited understanding of immune responses required to protect against HIV acquisition. Nonetheless, valuable knowledge accrued from numerous basic and translational science research studies and vaccine trials has provided insight into the structural biology of the virus, immunogen design and novel vaccine delivery systems that will likely constitute an effective vaccine. Furthermore, stakeholders now appreciate the daunting scientific challenges of developing an effective HIV vaccine, hence the increased advocacy for collaborative efforts among academic research scientists, governments, pharmaceutical industry, philanthropy, and regulatory entities. In this review, we highlight the history of HIV vaccine development efforts, highlighting major challenges and future directions.
Collapse
Affiliation(s)
- Tiza Ng'uni
- KwaZulu-Natal Research Institute for Tuberculosis and HIV (K-RITH), Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Caroline Chasara
- KwaZulu-Natal Research Institute for Tuberculosis and HIV (K-RITH), Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Zaza M Ndhlovu
- KwaZulu-Natal Research Institute for Tuberculosis and HIV (K-RITH), Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa.,Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA, United States
| |
Collapse
|
4
|
HIV-vaccines: lessons learned and the way forward. ASIAN BIOMED 2018. [DOI: 10.2478/abm-2010-0090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Abstract
A safe and efficacious preventive HIV vaccine, as part of a comprehensive prevention program, remains among the highest public health priorities. It would be the best tool that could reduce the spread of HIV significantly in the long run. Current AIDS vaccine candidates are unable to induce neutralizing antibodies against primary HIV isolates or only to a very limited and narrow extent, representing a major obstacle in the development of an efficacious HIV vaccine. Clinical efforts have mainly focused on T-cell vaccines such as DNA and various recombinant vectors alone or in prime-boost regimens. The Merck Ad5 vaccine not only failed to show efficacy but also was associated with increased risk of HIV acquisition in vaccinees in a Phase IIb trial. While gp120 alone was not efficacious, the ALVAC prime and gp120 boost regimen showed 31% efficacy in a Phase III trial in Thailand. These contrasting results illustrate the limitations of available laboratory assays to assess the vaccine-induced immune responses and the lack of understanding of immune correlates of protection. Efforts should therefore focus on developing vaccine candidates inducing broadly neutralizing antibodies. Similarly, new vector strategies such as replicating vectors should be explored to induce strong and broad T-cell responses in the systemic and mucosal compartments. Innovation in immune assay development and testing algorithms is critically needed. The standardization of more relevant and predictive non-human primate models for immunogenicity and efficacy studies will contribute to better and faster vaccine assessment. HIV vaccine development requires innovative ideas and a sustained long-term commitment of the scientific community, civil society, politicians, and donors and participants for clinical research.
Collapse
|
5
|
Dinges W, Girard PM, Podzamczer D, Brockmeyer NH, García F, Harrer T, Lelievre JD, Frank I, Colin De Verdière N, Yeni GP, Ortega Gonzalez E, Rubio R, Clotet Sala B, DeJesus E, Pérez-Elias MJ, Launay O, Pialoux G, Slim J, Weiss L, Bouchaud O, Felizarta F, Meurer A, Raffi F, Esser S, Katlama C, Koletar SL, Mounzer K, Swindells S, Baxter JD, Schneider S, Chas J, Molina JM, Koutsoukos M, Collard A, Bourguignon P, Roman F. The F4/AS01B HIV-1 Vaccine Candidate Is Safe and Immunogenic, But Does Not Show Viral Efficacy in Antiretroviral Therapy-Naive, HIV-1-Infected Adults: A Randomized Controlled Trial. Medicine (Baltimore) 2016; 95:e2673. [PMID: 26871794 PMCID: PMC4753889 DOI: 10.1097/md.0000000000002673] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The impact of the investigational human immunodeficiency virus type 1 (HIV-1) F4/AS01B vaccine on HIV-1 viral load (VL) was evaluated in antiretroviral therapy (ART)-naive HIV-1 infected adults.This phase IIb, observer-blind study (NCT01218113), included ART-naive HIV-1 infected adults aged 18 to 55 years. Participants were randomized to receive 2 (F4/AS01B_2 group, N = 64) or 3 (F4/AS01B_3 group, N = 62) doses of F4/AS01B or placebo (control group, N = 64) at weeks 0, 4, and 28. Efficacy (HIV-1 VL, CD4 T-cell count, ART initiation, and HIV-related clinical events), safety, and immunogenicity (antibody and T-cell responses) were evaluated during 48 weeks.At week 48, based on a mixed model, no statistically significant difference in HIV-1 VL change from baseline was demonstrated between F4/AS01B_2 and control group (0.073 log10 copies/mL [97.5% confidence interval (CI): -0.088; 0.235]), or F4/AS01B_3 and control group (-0.096 log10 copies/mL [97.5% CI: -0.257; 0.065]). No differences between groups were observed in HIV-1 VL change, CD4 T-cell count, ART initiation, or HIV-related clinical events at intermediate timepoints. Among F4/AS01B recipients, the most frequent solicited symptoms were pain at injection site (252/300 doses), fatigue (137/300 doses), myalgia (105/300 doses), and headache (90/300 doses). Twelve serious adverse events were reported in 6 participants; 1 was considered vaccine-related (F4/AS01B_2 group: angioedema). F4/AS01B induced polyfunctional F4-specific CD4 T-cells, but had no significant impact on F4-specific CD8 T-cell and anti-F4 antibody levels.F4/AS01B had a clinically acceptable safety profile, induced F4-specific CD4 T-cell responses, but did not reduce HIV-1 VL, impact CD4 T-cells count, delay ART initiation, or prevent HIV-1 related clinical events.
Collapse
Affiliation(s)
- Warren Dinges
- From the Seattle Travel and Preventive Medicine, Seattle Infectious Disease Clinic, Seattle, WA, USA (WD); Service des Maladies Infectieuses, Hôpital Saint Antoine, Assistance Publique Hôpitaux de Paris; and INSERM, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France (P-MG); HIV Unit, Infectious Disease Service, Hospital Universitari de Bellvitge, L'Hospitalet, 08907 Barcelona, Spain (DP); Department of Dermatology, Venerology, and Allergology, St. Josef-Hospital, Ruhr-Universität Bochum, Bochum, Germany (NHB); Hospital Clínic, IDIBAPS, University of Barcelona, Barcelona, Spain (FG); Department of Internal Medicine 3, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Germany (TH); Service d'Immunologie Clinique, Hôpital Henri Mondor, Créteil, France (J-DL); University of Pennsylvania, Philadelphia, PA, USA (IF); Service des Maladies Infectieuses et Tropicales, Hôpital Saint Louis, University of Paris Diderot Paris 7, Sorbonne Paris Cité and INSERM U941 (NCDV, J-MM); Hôpital Bichat Claude Bernard, Service des Maladies Infectieuses et Tropicales A, Paris, France (G-PY); Servicio de Enfermedades Infecciosas, Hospital General Universitario de Valencia, Valencia (EOG); Servicio de Enfermedades Infecciosas, Hospital 12 De Octubre, Madrid, Spain (RR); IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Uvic-UCC, Barcelona, Spain (BCS); Orlando Immunology Center, Orlando, FL, USA (EDS); Servicio de Enfermedades Infecciosas, Hospital Ramón Y Cajal, IRYCIS Madrid, Spain (MJPE); Université Paris Descartes, Sorbonne Paris Cité, Inserm, CIC 1417 and F-CRIN, Innovative Clinical Research Network in Vaccinology (I-REIVAC); and Assistance Publique Hôpitaux de Paris, Hôpital Cochin (OL); Maladies Infectieuses et Tropicales Co-infections, Hôpital Tenon, Paris, France (GP, JC); Saint Michael's Medical Center, Newark, NJ, USA (JS); Service d'immunologie Clinique, Hôpital Européen Georges Pompidou, Paris, France (LW); Service des Maladie Infectieuses et Tropicales, Hôpital Avicenne, Bobigny, France (OB); Private practice, Bakersfield, CA, USA (FF); Zentrum für Innere Medizin und Infektiologie, Praxis, München, Germany (AM); CMIT, 46 Rue Henri Huchard, Paris, France (FR); HIV Ambulanz, Klinik für Dermatologie, Uniklinikum Essen, Essen, Germany (SE); Service des Maladies Infectieuses et Tropicales, Hôpital de la Pitié-Salpêtrière, Paris, France (CK); The Ohio State University, Division of Infectious Diseases, Columbus, OH (SLK); Philadelphia FIGHT, Philadelphia, PA (KM); University of Nebraska Medical Center, Omaha, NE (SS); Cooper University Hospital, Cooper Medical School of Rowan University, Camden, NJ (JDB); Living Hope Clinical Foundation, Long Beach, CA, USA (SS); and GSK Vaccines, Wavre/Rixensart, Belgium (MK, AC, PB, FR)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Leroux-Roels G, Bourguignon P, Willekens J, Janssens M, Clement F, Didierlaurent AM, Fissette L, Roman F, Boutriau D. Immunogenicity and safety of a booster dose of an investigational adjuvanted polyprotein HIV-1 vaccine in healthy adults and effect of administration of chloroquine. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2014; 21:302-11. [PMID: 24391139 PMCID: PMC3957681 DOI: 10.1128/cvi.00617-13] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 12/21/2013] [Indexed: 11/20/2022]
Abstract
This phase II study evaluated the effect of chloroquine on the specific CD8(+) T-cell responses to and the safety of a booster dose of investigational human immunodeficiency virus type 1 (HIV-1) F4/AS01(B) vaccine containing 10 μg of recombinant fusion protein (F4) adjuvanted with the AS01(B) adjuvant system. Healthy adults aged 21 to 41 years, primed 3 years before with two F4/AS01(B) doses containing 10 or 30 μg of F4 (ClinicalTrials.gov registration number NCT00434512), were randomized (1:1) to receive the F4/AS01(B) booster administered alone or 2 days after chloroquine (300 mg). F4-specific CD8(+)/CD4(+) T-cell responses were characterized by intracellular cytokine staining and lymphoproliferation assays and anti-F4 antibodies by enzyme-linked immunosorbent assays (ELISAs). No effect of chloroquine on CD4(+)/CD8(+) T-cell and antibody responses and no vaccine effect on CD8(+) T-cell responses (cytokine secretion or proliferation) were detected following F4/AS01(B) booster administration. In vitro, chloroquine had a direct inhibitory effect on AS01(B) adjuvant properties; AS01-induced cytokine production decreased upon coincubation of cells with chloroquine. In the pooled group of participants primed with F4/AS01(B) containing 10 μg of F4, CD4(+) T-cell and antibody responses induced by primary vaccination persisted for at least 3 years. The F4/AS01(B) booster induced strong F4-specific CD4(+) T-cell responses, which persisted for at least 6 months with similar frequencies and polyfunctional phenotypes as following primary vaccination, and high anti-F4 antibody concentrations, reaching higher levels than those following primary vaccination. The F4/AS01(B) booster had a clinically acceptable safety and reactogenicity profile. An F4/AS01(B) booster dose, administered alone or after chloroquine, induced robust antibody and F4-specific CD4(+) T-cell responses but no significant CD8(+) T-cell responses (cytokine secretion or proliferation) in healthy adults. (This study has been registered at ClinicalTrials.gov under registration number NCT00972725).
Collapse
|
7
|
Louz D, Bergmans HE, Loos BP, Hoeben RC. Animal models in virus research: their utility and limitations. Crit Rev Microbiol 2012; 39:325-61. [PMID: 22978742 DOI: 10.3109/1040841x.2012.711740] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Viral diseases are important threats to public health worldwide. With the number of emerging viral diseases increasing the last decades, there is a growing need for appropriate animal models for virus studies. The relevance of animal models can be limited in terms of mimicking human pathophysiology. In this review, we discuss the utility of animal models for studies of influenza A viruses, HIV and SARS-CoV in light of viral emergence, assessment of infection and transmission risks, and regulatory decision making. We address their relevance and limitations. The susceptibility, immune responses, pathogenesis, and pharmacokinetics may differ between the various animal models. These complexities may thwart translating results from animal experiments to the humans. Within these constraints, animal models are very informative for studying virus immunopathology and transmission modes and for translation of virus research into clinical benefit. Insight in the limitations of the various models may facilitate further improvements of the models.
Collapse
Affiliation(s)
- Derrick Louz
- National Institute for Public Health and the Environment (RIVM), GMO Office , Bilthoven , The Netherlands
| | | | | | | |
Collapse
|
8
|
Kurihara K, Takahara Y, Nomura T, Ishii H, Iwamoto N, Takahashi N, Inoue M, Iida A, Hara H, Shu T, Hasegawa M, Moriya C, Matano T. Immunogenicity of repeated Sendai viral vector vaccination in macaques. Microbes Infect 2012; 14:1169-76. [PMID: 22884717 DOI: 10.1016/j.micinf.2012.07.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Revised: 06/21/2012] [Accepted: 07/21/2012] [Indexed: 11/29/2022]
Abstract
Induction of durable cellular immune responses by vaccination is an important strategy for the control of persistent pathogen infection. Viral vectors are promising vaccine tools for eliciting antigen-specific T-cell responses. Repeated vaccination may contribute to durable memory T-cell induction, but anti-vector antibodies could be an obstacle to its efficacy. We previously developed a Sendai virus (SeV) vector vaccine and showed the potential of this vector for efficient T-cell induction in macaques. Here, we examined whether repeated SeV vector vaccination with short intervals can enhance antigen-specific CD8(+) T-cell responses. Four rhesus macaques possessing the MHC-I haplotype 90-120-Ia were immunized three times with intervals of three weeks. For the vaccination, we used replication-defective F-deleted SeV vectors inducing CD8(+) T-cell responses specific for simian immunodeficiency virus Gag(206-216) and Gag(241-249), which are dominant epitopes restricted by 90-120-Ia-derived MHC-I molecules. All four animals showed higher Gag(206-216)-specific and Gag(241-249)-specific CD8(+) T-cell responses after the third vaccination than those after the first vaccination, indicating enhancement of antigen-specific CD8(+) T-cell responses by the second/third SeV vector vaccination even with short intervals. These results suggest that repeated SeV vector vaccination can contribute to induction of efficient and durable T-cell responses.
Collapse
Affiliation(s)
- Kyoko Kurihara
- AIDS Research Center, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Moriya C, Horiba S, Kurihara K, Kamada T, Takahara Y, Inoue M, Iida A, Hara H, Shu T, Hasegawa M, Matano T. Intranasal Sendai viral vector vaccination is more immunogenic than intramuscular under pre-existing anti-vector antibodies. Vaccine 2011; 29:8557-63. [PMID: 21939708 DOI: 10.1016/j.vaccine.2011.09.028] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Revised: 09/05/2011] [Accepted: 09/08/2011] [Indexed: 11/16/2022]
Abstract
Viral vectors are promising vaccine tools for eliciting potent cellular immune responses. Pre-existing anti-vector antibodies, however, can be an obstacle to their clinical use in humans. We previously developed a Sendai virus (SeV) vector vaccine and showed the potential of this vector for efficient CD8(+) T-cell induction in macaques. Here, we investigated the immunogenicity of SeV vector vaccination in the presence of anti-SeV antibodies. We compared antigen-specific CD8(+) T-cell responses after intranasal or intramuscular immunization with a lower dose (one-tenth of that in our previous studies) of SeV vector expressing simian immunodeficiency virus Gag antigen (SeV-Gag) between naive and pre-SeV-infected cynomolgus macaques. Intranasal SeV-Gag immunization efficiently elicited Gag-specific CD8(+) T-cell responses not only in naive but also in pre-SeV-infected animals. In contrast, intramuscular SeV-Gag immunization induced Gag-specific CD8(+) T-cell responses efficiently in naive but not in pre-SeV-infected animals. These results indicate that both intranasal and intramuscular SeV administrations are equivalently immunogenic in the absence of anti-SeV antibodies, whereas intranasal SeV vaccination is more immunogenic than intramuscular in the presence of anti-SeV antibodies. It is inferred from a recent report investigating the prevalence of anti-SeV antibodies in humans that SeV-specific neutralizing titers in more than 70% of people are no more than those at the SeV-Gag vaccination in pre-SeV-infected macaques in the present study. Taken together, this study implies the potential of intranasal SeV vector vaccination to induce CD8(+) T-cell responses even in humans, suggesting a rationale for proceeding to a vaccine clinical trial using this vector.
Collapse
Affiliation(s)
- Chikaya Moriya
- The Institute of Medical Science, The University of Tokyo, Shirokanedai, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Socioecological influences on community involvement in HIV vaccine research. Vaccine 2011; 29:6136-43. [PMID: 21722689 DOI: 10.1016/j.vaccine.2011.06.082] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Revised: 05/31/2011] [Accepted: 06/14/2011] [Indexed: 11/21/2022]
Abstract
OBJECTIVE This study investigated socioecological factors influencing HIV vaccine research participation among communities living in geographic areas with high HIV prevalence and high poverty rates. METHODS We surveyed a sample of 453 adults ≤18 years from areas of high poverty and high HIV prevalence in metro Atlanta and differentiated the effects of individual-, social/organizational-, and community-level characteristics on participation in HIV vaccine research via multilevel modeling techniques that incorporated questionnaire, program, and census data. RESULTS Models that adjusted for both individual-level covariates (such as race, gender, attitudes, and beliefs concerning HIV research), social/organizational- and community-level factors such as local HIV prevalence rates, revealed that the extent of HIV prevention-related programs and services in census tracts contributed to individuals' likelihood of participation in an HIV vaccine study. Additionally, neighborhood-based organizations offering HIV medical and treatment programs, support groups, and services (e.g., food, shelter, and clothing) encourage greater HIV vaccine research participation. CONCLUSIONS The findings support the hypothesis that community-level factors facilitate participation in HIV vaccine research independent of both individual- and social/organizational-level factors.
Collapse
|
11
|
Excler JL, Parks CL, Ackland J, Rees H, Gust ID, Koff WC. Replicating viral vectors as HIV vaccines: summary report from the IAVI-sponsored satellite symposium at the AIDS vaccine 2009 conference. Biologicals 2011; 38:511-21. [PMID: 20537552 DOI: 10.1016/j.biologicals.2010.03.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2010] [Accepted: 03/29/2010] [Indexed: 01/30/2023] Open
Abstract
In October 2009, The International AIDS Vaccine Initiative (IAVI) convened a satellite symposium entitled 'Replicating Viral Vectors for use in AIDS Vaccines' at the AIDS Vaccine 2009 Conference in Paris. The purpose of the symposium was to gather together researchers, representatives from regulatory agencies, and vaccine developers to discuss issues related to advancement of replication-competent viral vector- based HIV vaccines into clinical trials. The meeting introduced the rationale for accelerating the development of replicating viral vectors for use as AIDS vaccines. It noted that the EMEA recently published draft guidelines that are an important first step in providing guidance for advancing live viral vectors into clinical development. Presentations included case studies and development challenges for viral vector-based vaccine candidates. These product development challenges included cell substrates used for vaccine manufacturing, the testing needed to assess vaccine safety, conducting clinical trials with live vectors, and assessment of vaccination risk versus benefit. More in depth discussion of risk and benefit highlighted the fact that AIDS vaccine efficacy trials must be conducted in the developing world where HIV incidence is greatest and how inequities in global health dramatically influence the political and social environment in developing countries.
Collapse
Affiliation(s)
- J L Excler
- International AIDS Vaccine Initiative, 110 William Street, 27th Floor, New York, NY 10038-3901, USA
| | | | | | | | | | | |
Collapse
|
12
|
Abstract
PURPOSE OF REVIEW Following the evidence that T-cell responses are crucial in the control of HIV-1 infection, vaccines targeting T-cell responses were tested in recent clinical trials. However, these vaccines showed a lack of efficacy. This review attempts to define the qualitative and quantitative features that are desirable for T-cell-induced responses by vaccines. We also describe strategies that could lead to achievement of this goal. RECENT FINDINGS Using the yellow fever vaccine as a benchmark of an efficient vaccine, recent studies identified factors of immune protection and more importantly innate immune pathways needed for the establishment of long-term protective adaptive immunity. SUMMARY To prevent or control HIV-1 infection, a vaccine must induce efficient and persistent antigen-specific T cells endowed with mucosal homing capacity. Such cells should have the capability to counteract HIV-1 diversity and its rapid spread from the initial site of infection. To achieve this goal, the activation of a diversified innate immune response is critical. New systems biology approaches will provide more precise correlates of immune protection that will pave the way for new approaches in T-cell-based vaccines.
Collapse
|
13
|
Abstract
Microbial pathogens have developed complex and efficient ways of counteracting and evading innate and adaptive immune mechanisms. The strategies used by pathogens determine strongly the type of immune response a vaccine should elicit and how the vaccine should be formulated. Improved knowledge of immune response mechanisms has brought successes in the development of vaccines that protect against challenging pathogens as well as vaccines that can be used in immunocompromised and elderly populations. This includes the production of highly purified antigens that provide a better reactogenicity and safety profile than some of the early whole-pathogen vaccines. Successful attempts to improve antigen purity, however, can result in weakened immunogenicity. The search for approaches to overcome this has led to new technologies, such as live vector vaccines, DNA vaccines and novel adjuvant formulations, which have been based on growing knowledge of the interplay between innate and adaptive immune systems and the central role played by antigen-presenting cells. Of these technologies, one of the most promising to date is based on the use of innovative adjuvants combined with careful antigen selection. Vaccine design has therefore become more tailored, and in turn has opened up the potential of extending its application in immunotherapies to tackle diseases such as cancer, Alzheimer disease and immune-mediated disorders.
Collapse
Affiliation(s)
- Fred Zepp
- University Medical Center, Department of Pediatrics, Mainz, Germany.
| |
Collapse
|
14
|
Leroux-Roels I, Koutsoukos M, Clement F, Steyaert S, Janssens M, Bourguignon P, Cohen K, Altfeld M, Vandepapelière P, Pedneault L, McNally L, Leroux-Roels G, Voss G. Strong and persistent CD4+ T-cell response in healthy adults immunized with a candidate HIV-1 vaccine containing gp120, Nef and Tat antigens formulated in three Adjuvant Systems. Vaccine 2010; 28:7016-24. [DOI: 10.1016/j.vaccine.2010.08.035] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2010] [Revised: 07/25/2010] [Accepted: 08/03/2010] [Indexed: 10/19/2022]
|
15
|
Andanda P. The role of law in the regulation of HIV-vaccine research in South Africa and Kenya. AFRICAN JOURNAL OF AIDS RESEARCH : AJAR 2010; 9:249-259. [PMID: 25860629 DOI: 10.2989/16085906.2010.530178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Law is an important regulatory mechanism, particularly for creating an enabling research environment. However, the manner in which law addresses issues related to medical research, and HIV-vaccine research in particular, is at times inadequate and of great concern to the stakeholders involved in such research. The challenges for law as a regulatory mechanism in this regard are twofold: the complexity of issues related to HIV-vaccine research, and the apparent disconnection between the legal and ethical frameworks that are applied in the regulation of these issues. This article analyses the extent to which these challenges have been addressed in South Africa and Kenya. Especially, it highlights the lessons that can be drawn from the two countries in establishing a suitable ethical-legal framework for HIV-vaccine research.
Collapse
Affiliation(s)
- Pamela Andanda
- a University of the Witwatersrand , School of Law , Private Bag 3 , Johannesburg , 2050 , South Africa
| |
Collapse
|
16
|
Frew PM, Archibald M, Diallo DD, Hou SI, Horton T, Chan K, Mulligan MJ, del Rio C. An extended model of reasoned action to understand the influence of individual- and network-level factors on African Americans' participation in HIV vaccine research. PREVENTION SCIENCE : THE OFFICIAL JOURNAL OF THE SOCIETY FOR PREVENTION RESEARCH 2010; 11:207-18. [PMID: 20012200 PMCID: PMC2858782 DOI: 10.1007/s11121-009-0162-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In the United States, the number and proportion of HIV/AIDS cases among black/African Americans continue to highlight the need for new biomedical prevention interventions, including an HIV vaccine, microbicide, or new antiretroviral (ARV) prevention strategies such as pre-exposure prophylaxis (PrEP) to complement existing condom usage, harm reduction methods, and behavioral change strategies to stem the HIV epidemic. Although black/African Americans are disproportionately impacted by HIV/AIDS, their participation in HIV clinical research continues to have unique challenges. We theorize that interaction among multilevel factors creates ideal alignment for minority participation in HIV clinical studies. Thus, we initially set out to test an extended model of reasoned action with 362 participants to understand the interplay of sociopsychological and network-level considerations influencing minority participation in HIV prevention research efforts. In this study, we linked the intrapersonal dimensions of attitudes, beliefs, and normative concerns to community-level components, appraisal of involvement with the clinical research organization, an entity which operates within a networked structure of community partner agencies, and identification with coalition advocacy aims. Various participatory outcomes were explored including involvement in future HIV vaccine community functions, participation in community promotion of HIV vaccine research, and community mobilization. Three-stage least squares estimates indicated similar findings across three models. Significant effects demonstrate the importance of positive attitudes toward HIV vaccine research, favorable health research beliefs, perceived social support for participation, HIV/AIDS issue engagement, and perceived relevance of the clinical research site's mission and values. Identification of these nuanced pathway effects provides implications for tailored community program development.
Collapse
Affiliation(s)
- Paula M Frew
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA.
| | | | | | | | | | | | | | | |
Collapse
|
17
|
The emerging shape of a global HIV research agenda: how partnerships between Northern and Southern researchers are addressing questions relevant to both. Curr Opin HIV AIDS 2009; 3:521-5. [PMID: 19373015 DOI: 10.1097/coh.0b013e3283025e3d] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
PURPOSE OF REVIEW In this review I will narrate and discuss recent North-South health research partnerships and how such partnerships are of mutual benefit to both. RECENT DEVELOPMENTS Globalization has shrunk the world into a virtual single community in which actions of individuals may have global repercussion. The ever-improving accessibility to fast communications through both transportation and knowledge and information exchange is making this virtual community even smaller. The current large global burden of diseases of poverty, made worse by the HIV/AIDS pandemic is a major concern for all. Although the South bears the brunt of this burden and is disproportionately adversely affected, the ill effects are global. Among these effects, include global poverty, development constraints and insecurity. It is no wonder that scientists from both north and south are working together to mitigate the effects of this scourge for the mutual benefit of all. SUMMARY Despite a period of 25 years since the onset of the HIV/AIDS pandemic many challenges still abound. A preventive vaccine is still illusive and care and prevention programmes not universal. To mitigate this several global initiatives and North-South partnerships are working together for the common good. Such collaborations seen in academic settings, research institutions, networks of excellence and the private sector engage in research, training and service provision. This cooperation includes discovery and evaluations of HIV vaccine candidates as well as treatment and prevention methods.
Collapse
|
18
|
Galárraga O, Colchero MA, Wamai RG, Bertozzi SM. HIV prevention cost-effectiveness: a systematic review. BMC Public Health 2009; 9 Suppl 1:S5. [PMID: 19922689 PMCID: PMC2779507 DOI: 10.1186/1471-2458-9-s1-s5] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND After more than 25 years, public health programs have not been able to sufficiently reduce the number of new HIV infections. Over 7,000 people become infected with HIV every day. Lack of convincing evidence of cost-effectiveness (CE) may be one of the reasons why implementation of effective programs is not occurring at sufficient scale. This paper identifies, summarizes and critiques the CE literature related to HIV-prevention interventions in low- and middle-income countries during 2005-2008. METHODS Systematic identification of publications was conducted through several methods: electronic databases, internet search of international organizations and major funding/implementing agencies, and journal browsing. Inclusion criteria included: HIV prevention intervention, year for publication (2005-2008), setting (low- and middle-income countries), and CE estimation (empirical or modeling) using outcomes in terms of cost per HIV infection averted and/or cost per disability-adjusted life year (DALY) or quality-adjusted life year (QALY). RESULTS We found 21 distinct studies analyzing the CE of HIV-prevention interventions published in the past four years (2005-2008). Seventeen CE studies analyzed biomedical interventions; only a few dealt with behavioral and environmental/structural interventions. Sixteen studies focused on sub-Saharan Africa, and only a handful on Asia, Latin America and Eastern Europe. Many HIV-prevention interventions are very cost effective in absolute terms (using costs per DALY averted), and also in country-specific relative terms (in cost per DALY measured as percentage of GDP per capita). CONCLUSION There are several types of interventions for which CE studies are still not available or insufficient, including surveillance, abstinence, school-based education, universal precautions, prevention for positives and most structural interventions. The sparse CE evidence available is not easily comparable; thus, not very useful for decision making. More than 25 years into the AIDS epidemic and billions of dollars of spending later, there is still much work to be done both on costs and effectiveness to adequately inform HIV prevention planning.
Collapse
Affiliation(s)
- Omar Galárraga
- Center for Evaluation Research and Surveys, Mexican School of Public Health/National Institute of Public Health (INSP), Av. Universidad 655, Cuernavaca, Mexico CP 62508
- Haas School of Business, University of California, Berkeley, CA, USA
| | - M Arantxa Colchero
- Center for Evaluation Research and Surveys, Mexican School of Public Health/National Institute of Public Health (INSP), Av. Universidad 655, Cuernavaca, Mexico CP 62508
| | - Richard G Wamai
- Department of African-American Studies, Northeastern University, Boston, MA, USA; Harvard School of Public Health, Cambridge, MA, USA; Nairobi University, Department of Community Health, Nairobi, Kenya
| | - Stefano M Bertozzi
- Center for Evaluation Research and Surveys, Mexican School of Public Health/National Institute of Public Health (INSP), Av. Universidad 655, Cuernavaca, Mexico CP 62508
- Haas School of Business, University of California, Berkeley, CA, USA
- Center for Economic Teaching and Research (CIDE), Mexico City, Mexico
| |
Collapse
|
19
|
Ramanathan VD, Kumar M, Mahalingam J, Sathyamoorthy P, Narayanan PR, Solomon S, Panicali D, Chakrabarty S, Cox J, Sayeed E, Ackland J, Verlinde C, Vooijs D, Loughran K, Barin B, Lombardo A, Gilmour J, Stevens G, Smith MS, Tarragona-Fiol T, Hayes P, Kochhar S, Excler JL, Fast P. A Phase 1 study to evaluate the safety and immunogenicity of a recombinant HIV type 1 subtype C-modified vaccinia Ankara virus vaccine candidate in Indian volunteers. AIDS Res Hum Retroviruses 2009; 25:1107-16. [PMID: 19943789 DOI: 10.1089/aid.2009.0096] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
A recombinant modified vaccinia Ankara virus vaccine candidate (TBC-M4) expressing HIV-1 subtype C env, gag, tat-rev, and nef-RT genes was tested in a randomized, double-blind, dose escalation Phase I trial in 32 HIV-uninfected healthy volunteers who received three intramuscular injections of TBC-M4 at 0, 1, and 6 months of 5 x 10(7) plaque-forming units (pfu) (low dosage, LD) (n = 12) or 2.5 x 10(8) pfu (high dosage, HD) (n = 12) or placebo (n = 8). Local and systemic reactogenicity was experienced by approximately 67% and 83% of vaccine recipients, respectively. The reactogenicity events were mostly mild in severity. Severe but transient systemic reactogenicity was seen in one volunteer of the HD group. No vaccine-related serious adverse events or events suggesting perimyocarditis were seen. A higher frequency of local reactogenicity events was observed in the HD group. Cumulative HIV-specific IFN-gamma ELISPOT responses were detected in frozen PBMCs from 9/11 (82%), 12/12 (100%), and 1/8 (13%) volunteers after the third injection of the LD, HD, and placebo groups, respectively. Most of the responses were to gag and env proteins (maximum of 430 SFU/10(6) PBMCs) persisting across multiple time points. HIV-specific ELISA antibody responses were detected in 10/11, 12/12, and 0/8 volunteers post-third vaccination, in the LD, HD, and placebo groups, respectively. No neutralizing activity against HIV-1 subtype C isolates was detected. TBC-M4 appears to be generally safe and well-tolerated. The immune response detected was dose dependent, modest in magnitude, and directed mostly to env and gag proteins, suggesting further evaluation of this vaccine in a prime-boost regimen.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Josephine Cox
- International AIDS Vaccine Initiative, New York, New York 10038
| | - Eddy Sayeed
- International AIDS Vaccine Initiative, New York, New York 10038
| | - James Ackland
- International AIDS Vaccine Initiative, New York, New York 10038
| | - Carl Verlinde
- International AIDS Vaccine Initiative, New York, New York 10038
| | - Dani Vooijs
- International AIDS Vaccine Initiative, New York, New York 10038
| | | | - Burc Barin
- EMMES Corporation, Rockville Maryland 20850
| | - Angela Lombardo
- International AIDS Vaccine Initiative, New York, New York 10038
| | - Jill Gilmour
- IAVI Human Immunology Laboratory, Imperial College, London, UK
| | | | | | | | | | | | | | - Patricia Fast
- International AIDS Vaccine Initiative, New York, New York 10038
| |
Collapse
|
20
|
Surman SL, Sealy R, Jones BG, Hurwitz JL. HIV-1 vaccine design: harnessing diverse lymphocytes to conquer a diverse pathogen. HUMAN VACCINES 2009; 5:268-71. [PMID: 19684481 DOI: 10.4161/hv.5.4.7706] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In the fall of 2007, the HIV-1 research field received news that their front-runner vaccine was not protective. In response to this disappointment, scientists are now reviewing the intricacies of the immune response toward HIV-1 to develop new and better strategies for vaccine development. Decades ago, researchers recognized the impressive amino acid and carbohydrate diversity of HIV-1, and the associated obstacles to vaccine development. At first glance, the diversity and other unique features of HIV-1 may seem insurmountable, but attention to vaccine successes in other fields serves to renew optimism. The newly-licensed rotavirus and papillomavirus cocktail vaccines remind scientists that diverse pathogens can be conquered and that the chronic nature of a virus infection need not thwart successful vaccine design. Here we describe current efforts to gain insights from other vaccine fields and to adopt a cocktail vaccine approach for the prevention of HIV-1 infections in humans.
Collapse
Affiliation(s)
- Sherri L Surman
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | | | | |
Collapse
|
21
|
Engram JC, Dunham RM, Makedonas G, Vanderford TH, Sumpter B, Klatt NR, Ratcliffe SJ, Garg S, Paiardini M, McQuoid M, Altman JD, Staprans SI, Betts MR, Garber DA, Feinberg MB, Silvestri G. Vaccine-induced, simian immunodeficiency virus-specific CD8+ T cells reduce virus replication but do not protect from simian immunodeficiency virus disease progression. THE JOURNAL OF IMMUNOLOGY 2009; 183:706-17. [PMID: 19542473 DOI: 10.4049/jimmunol.0803746] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Our limited understanding of the interaction between primate lentiviruses and the host immune system complicates the design of an effective HIV/AIDS vaccine. To identify immunological correlates of protection from SIV disease progression, we immunized two groups of five rhesus macaques (RMs) with either modified vaccinia Ankara (MVA) or MVADeltaudg vectors that expressed SIVmac239 Gag and Tat. Both vectors raised a SIV-specific CD8(+) T cell response, with a magnitude that was greater in mucosal tissues than in peripheral blood. After challenge with SIVmac239, all vaccinated RMs showed mucosal and systemic CD8(+) T cell recall responses that appeared faster and were of greater magnitude than those in five unvaccinated control animals. All vaccinated RMs showed a approximately 1-log lower peak and early set-point SIV viral load than the unvaccinated animals, and then, by 8 wk postchallenge, exhibited levels of viremia similar to the controls. We observed a significant direct correlation between the magnitude of postchallenge SIV-specific CD8(+) T cell responses and SIV viral load. However, vaccinated RMs showed no protection from either systemic or mucosal CD4(+) T cell depletion and no improved survival. The observation that vaccine-induced, SIV-specific CD8(+) T cells that partially control SIVmac239 virus replication fail to protect from immunological or clinical progression of SIV infection underscores both the complexity of AIDS pathogenesis and the challenges of properly assessing the efficacy of candidate AIDS vaccines.
Collapse
Affiliation(s)
- Jessica C Engram
- Department of Pathology, University of Pennsylvania, Philadelphia, 19104, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Lau CY, Stansbury JP, Gust DA, Kafaar Z. Social and behavioral science in HIV vaccine trials: a gap assessment of the literature. Expert Rev Vaccines 2009; 8:179-90. [PMID: 19196198 DOI: 10.1586/14760584.8.2.179] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Social and behavioral science research is integral to the conduct of HIV vaccine trials, especially because the vaccine targets an infection laden with sensitive human issues. Although social and behavioral sciences have played a larger role in HIV vaccine clinical trials than other vaccine clinical trials to date, this role should be expanded. Fortunately, related publications, conference coverage and research proposals are on the rise; community engagement is receiving more attention during the earlier stages of product development; and collaboration between HIV vaccine scientists and social and behavioral scientists is being fostered. Greater attention to social and behavioral science issues could not only facilitate accrual, but also improve research efficiency and relevance. In this review, gaps in the literature on social and behavioral science issues in HIV vaccine clinical research, including barriers and facilitators to trial participation, enhancing feasibility of trial success, health systems, policy and monitoring social and behavioral issues, are identified and directions are suggested for filling those gaps. Development of a safe, efficacious and acceptable HIV vaccine will be nurtured by addressing the gaps through interdisciplinary collaborations.
Collapse
Affiliation(s)
- Chuen-Yen Lau
- Vaccine Clinical Research Branch, Vaccine Research Program, National Institute of Allergy and Infectious Disease, National Institutes of Health, 6700 B Rockledge, Room 5126, Bethesda, MD 20817, USA.
| | | | | | | |
Collapse
|
23
|
Optimization of human immunodeficiency virus gag expression by newcastle disease virus vectors for the induction of potent immune responses. J Virol 2008; 83:584-97. [PMID: 19004953 DOI: 10.1128/jvi.01443-08] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
One attractive strategy for the development of a human immunodeficiency virus (HIV) vaccine is the use of viral vectors with a proven safety profile and an absence of preexisting immunity in humans, such as Newcastle disease virus (NDV). Several NDV vaccine vectors have been generated, and their immunogenicities have been investigated with different animal models. However, a systematic study to evaluate the optimal insertion site of the foreign antigens into NDV that results in enhanced immune responses specific to the antigen has not yet been conducted. In this article, we describe the ability of NDV expressing HIV Gag to generate a Gag-specific immune response in mice. We also have determined the optimal insertion site into the NDV genome by generating recombinant NDV-HIVGag viruses in which HIV gag was located at different transcriptional positions throughout the NDV viral genome. All recombinant viruses were viable, grew to similar titers in embryonated chicken eggs, and expressed Gag in a stable manner. Our in vivo experiments revealed that higher HIV Gag protein expression positively correlates with an enhanced CD8(+) T-cell-mediated immune response and protective immunity against challenge with vaccinia virus expressing HIV Gag. We also inserted a codon-optimized version of HIV gag in the described best location, between the P and M genes. Virus expressing the codon-optimized version of HIV gag induced a higher expression of the protein and an enhanced immune response against HIV Gag in mice. These results indicate that strategies directed toward increasing antigen expression by NDV result in enhanced immunogenicity and vaccine efficacy.
Collapse
|
24
|
|
25
|
Abstract
The HIV/AIDS pandemic has become part of the contemporary global landscape. Few predicted its effect on mortality and morbidity or its devastating social and economic consequences, particularly in sub-Saharan Africa. Successful responses have addressed sensitive social factors surrounding HIV prevention, such as sexual behaviour, drug use, and gender equalities, countered stigma and discrimination, and mobilised affected communities; but such responses have been few and far between. Only in recent years has the international response to HIV prevention gathered momentum, mainly due to the availability of treatment with antiretroviral drugs, the recognition that the pandemic has both development and security implications, and a substantial increase in financial resources brought about by new funders and funding mechanisms. We now require an urgent and revitalised global movement for HIV prevention that supports a combination of behavioural, structural, and biomedical approaches and is based on scientifically derived evidence and the wisdom and ownership of communities.
Collapse
|
26
|
Schweizer A, Rusert P, Berlinger L, Ruprecht CR, Mann A, Corthésy S, Turville SG, Aravantinou M, Fischer M, Robbiani M, Amstutz P, Trkola A. CD4-specific designed ankyrin repeat proteins are novel potent HIV entry inhibitors with unique characteristics. PLoS Pathog 2008; 4:e1000109. [PMID: 18654624 PMCID: PMC2453315 DOI: 10.1371/journal.ppat.1000109] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2008] [Accepted: 06/24/2008] [Indexed: 11/18/2022] Open
Abstract
Here, we describe the generation of a novel type of HIV entry inhibitor using the recently developed Designed Ankyrin Repeat Protein (DARPin) technology. DARPin proteins specific for human CD4 were selected from a DARPin DNA library using ribosome display. Selected pool members interacted specifically with CD4 and competed with gp120 for binding to CD4. DARPin proteins derived in the initial selection series inhibited HIV in a dose-dependent manner, but showed a relatively high variability in their capacity to block replication of patient isolates on primary CD4 T cells. In consequence, a second series of CD4-specific DARPins with improved affinity for CD4 was generated. These 2nd series DARPins potently inhibit infection of genetically divergent (subtype B and C) HIV isolates in the low nanomolar range, independent of coreceptor usage. Importantly, the actions of the CD4 binding DARPins were highly specific: no effect on cell viability or activation, CD4 memory cell function, or interference with CD4-independent virus entry was observed. These novel CD4 targeting molecules described here combine the unique characteristics of DARPins-high physical stability, specificity and low production costs-with the capacity to potently block HIV entry, rendering them promising candidates for microbicide development.
Collapse
Affiliation(s)
- Andreas Schweizer
- Division of Infectious Diseases, University Hospital Zurich, Zurich, Switzerland
| | - Peter Rusert
- Division of Infectious Diseases, University Hospital Zurich, Zurich, Switzerland
| | - Livia Berlinger
- Division of Infectious Diseases, University Hospital Zurich, Zurich, Switzerland
| | - Claudia R. Ruprecht
- Division of Infectious Diseases, University Hospital Zurich, Zurich, Switzerland
| | - Axel Mann
- Division of Infectious Diseases, University Hospital Zurich, Zurich, Switzerland
| | - Stéphanie Corthésy
- Division of Infectious Diseases, University Hospital Zurich, Zurich, Switzerland
| | - Stuart G. Turville
- Center for Biomedical Research, Population Council, New York, New York, United States of America
| | - Meropi Aravantinou
- Center for Biomedical Research, Population Council, New York, New York, United States of America
| | - Marek Fischer
- Division of Infectious Diseases, University Hospital Zurich, Zurich, Switzerland
| | - Melissa Robbiani
- Center for Biomedical Research, Population Council, New York, New York, United States of America
| | | | - Alexandra Trkola
- Division of Infectious Diseases, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
27
|
Schellens IMM, Borghans JAM, Jansen CA, De Cuyper IM, Geskus RB, van Baarle D, Miedema F. Abundance of early functional HIV-specific CD8+ T cells does not predict AIDS-free survival time. PLoS One 2008; 3:e2745. [PMID: 18648514 PMCID: PMC2447878 DOI: 10.1371/journal.pone.0002745] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Accepted: 06/22/2008] [Indexed: 12/04/2022] Open
Abstract
Background T-cell immunity is thought to play an important role in controlling HIV infection, and is a main target for HIV vaccine development. HIV-specific central memory CD8+ and CD4+ T cells producing IFNγ and IL-2 have been associated with control of viremia and are therefore hypothesized to be truly protective and determine subsequent clinical outcome. However, the cause-effect relationship between HIV-specific cellular immunity and disease progression is unknown. We investigated in a large prospective cohort study involving 96 individuals of the Amsterdam Cohort Studies with a known date of seroconversion whether the presence of cytokine-producing HIV-specific CD8+ T cells early in infection was associated with AIDS-free survival time. Methods and Findings The number and percentage of IFNγ and IL-2 producing CD8+ T cells was measured after in vitro stimulation with an overlapping Gag-peptide pool in T cells sampled approximately one year after seroconversion. Kaplan-Meier survival analysis and Cox proportional hazard models showed that frequencies of cytokine-producing Gag-specific CD8+ T cells (IFNγ, IL-2 or both) shortly after seroconversion were neither associated with time to AIDS nor with the rate of CD4+ T-cell decline. Conclusions These data show that high numbers of functional HIV-specific CD8+ T cells can be found early in HIV infection, irrespective of subsequent clinical outcome. The fact that both progressors and long-term non-progressors have abundant T cell immunity of the specificity associated with low viral load shortly after seroconversion suggests that the more rapid loss of T cell immunity observed in progressors may be a consequence rather than a cause of disease progression.
Collapse
Affiliation(s)
| | - José A. M. Borghans
- Department of Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- Theoretical Biology/Bioinformatics, Utrecht University, Utrecht, The Netherlands
| | - Christine A. Jansen
- Department of Clinical Viro-Immunology, Sanquin Research and Landsteiner Laboratory of the Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Iris M. De Cuyper
- Department of Clinical Viro-Immunology, Sanquin Research and Landsteiner Laboratory of the Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Ronald B. Geskus
- Department of Research, Cluster, Infectious Diseases, Health Service of Amsterdam, Amsterdam, The Netherlands
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Debbie van Baarle
- Department of Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Frank Miedema
- Department of Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
28
|
Priddy FH, Brown D, Kublin J, Monahan K, Wright DP, Lalezari J, Santiago S, Marmor M, Lally M, Novak RM, Brown SJ, Kulkarni P, Dubey SA, Kierstead LS, Casimiro DR, Mogg R, DiNubile MJ, Shiver JW, Leavitt RY, Robertson MN, Mehrotra DV, Quirk E. Safety and immunogenicity of a replication-incompetent adenovirus type 5 HIV-1 clade B gag/pol/nef vaccine in healthy adults. Clin Infect Dis 2008; 46:1769-81. [PMID: 18433307 DOI: 10.1086/587993] [Citation(s) in RCA: 190] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The safety and immunogenicity of the MRK adenovirus type 5 human immunodeficiency virus type 1 clade B gag/pol/nef vaccine, a replication-incompetent adenovirus type 5-vectored vaccine designed to elicit cell-mediated immunity against conserved human immunodeficiency virus proteins, was assessed in a phase 1 trial. METHODS Healthy adults not infected with human immunodeficiency virus were enrolled in a multicenter, dose-escalating, blind, placebo-controlled study to evaluate a 3-dose homologous prime-boost regimen of the trivalent MRK adenovirus type 5 human immunodeficiency virus type 1 vaccine containing from 3 x 10(6) to 1 x 10(11) viral particles per 1-mL dose administered on day 1, during week 4 and during week 26. Adverse events were recorded for 29 days after each intradeltoid injection. The primary immunogenicity end point was the proportion of study participants with a positive unfractionated Gag-, Pol-, or Nef-specific interferon-gamma enzyme-linked immunosorbent spot response measured 4 weeks after administration of the last dose. RESULTS Of 259 randomized individuals, 257 (99%) received > or = 1 dose of vaccine or placebo and were included in the safety analyses. Enzyme-linked immunosorbent spot results were available for 217 study participants (84%) at week 30. No serious vaccine-related adverse events occurred. No study participant discontinued participation because of vaccine-related adverse events. The frequency of injection-site reactions was dose dependent. Vaccine doses of > or = 3 x 10(9) viral particles elicited positive enzyme-linked immunosorbent spot responses to > or = 1 vaccine component in > 60% of recipients. High baseline antibody titers against adenovirus type 5 diminished enzyme-linked immunosorbent spot responses at all doses except the 3 x 10(10) viral particle dose. CONCLUSIONS The vaccine was generally well tolerated and induced cell-mediated immune responses against human immunodeficiency virus type 1 peptides in most healthy adults. Despite these findings, vaccination in a proof-of-concept trial with use of this vaccine was discontinued because of lack of efficacy.
Collapse
Affiliation(s)
- Frances H Priddy
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
The discovery of RNA interference (RNAi) has resulted in a new class of biological agents that can specifically downmodulate HIV-1 gene expression. Delivery of these RNAi-based agents and the emergence of viral resistance present pressing issues in the use of RNAi in a genetic-based therapy for HIV-1. Here, we discuss a potential avenue around viral resistance and a targeted delivery scheme for treating HIV-1-infected individuals involving transcriptional gene silencing. Specifically, the use of small antisense RNAs targeted to the viral promoter regions and delivery by lentiviral-based mobilization-competent vectors expressing these promoter targeted RNAs may prove therapeutically relevant in a genetic therapy-based approach to treating HIV-1 infection.
Collapse
Affiliation(s)
- Anne-Marie W Turner
- Department of Molecular & Experimental Medicine and Kellogg School of Science & Technology, The Scripps Research Institute, 10550 N. Torrey Pines Road, MEM-115, La Jolla, CA 92037, USA
| | - Kevin V Morris
- Department of Molecular & Experimental Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Road, MEM-115, La Jolla, CA 92037, USA
| |
Collapse
|
30
|
Sekaly RP. The failed HIV Merck vaccine study: a step back or a launching point for future vaccine development? ACTA ACUST UNITED AC 2008; 205:7-12. [PMID: 18195078 PMCID: PMC2234358 DOI: 10.1084/jem.20072681] [Citation(s) in RCA: 307] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The world of human immunodeficiency virus (HIV) vaccines has suffered a baffling setback. The first trial of a vaccine designed to elicit strong cellular immunity has shown no protection against infection. More alarmingly, the vaccine appeared to increase the rate of HIV infection in individuals with prior immunity against the adenovirus vector used in the vaccine. A new study in this issue suggests that a different vaccine approach—using a DNA prime/poxvirus boost strategy—induces polyfunctional immune responses to an HIV immunogen. The disappointing results of the recent vaccine trial suggest that a more thorough assessment of vaccine-induced immune responses is urgently needed, and that more emphasis should be placed on primate models before efficacy trials are undertaken.
Collapse
Affiliation(s)
- Rafick-Pierre Sekaly
- Université de Montréal, CR-CHUM, Institut National de la Santé et de la Recherche Médicale U743, Montréal, Québec H2X1P1, Canada.
| |
Collapse
|