1
|
Hodgins B, Pillet S, Landry N, Ward BJ. Prime-pull vaccination with a plant-derived virus-like particle influenza vaccine elicits a broad immune response and protects aged mice from death and frailty after challenge. IMMUNITY & AGEING 2019; 16:27. [PMID: 31700523 PMCID: PMC6829930 DOI: 10.1186/s12979-019-0167-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 10/10/2019] [Indexed: 12/30/2022]
Abstract
Background Administered intramuscularly (IM), plant-derived, virus-like-particle (VLP) vaccines based on the influenza hemagglutinin (HA) protein elicit both humoral and cellular responses that can protect aged mice from lethal challenge. Unlike split virus vaccines, VLPs can be administered by different routes including intranasally (IN). We evaluated novel vaccine strategies such as prime-pull (IM boosted by IN) and multi-modality vaccination (IM and IN given simultaneously). We wished to determine if these approaches would provide better quality protection in old mice after less severe (borderline-lethal) challenge (ie: immunogenicity, frailty and survival). Results Survival rates were similar in all vaccinated groups. Antibody responses were modest in all groups but tended to be higher in VLP groups compared to inactivated influenza vaccine (IIV) recipients. All VLP groups had higher splenocyte T cell responses than the split virus group. Lung homogenate chemokine/cytokine levels and virus loads were lower in the VLP groups compared to IIV recipients 3 days after challenge (p < 0.05 for viral load vs all VLP groups combined). The VLP-vaccinated groups also had less weight loss and recovered more rapidly than the IIV recipients. There was limited evidence of an immunologic or survival advantage with IN delivery of the VLP vaccine. Conclusion Compared to IIV, the plant-derived VLP vaccine induced a broader immune response in aged mice (cellular and humoral) using either traditional (IM/IM) or novel schedules (multi-modality, prime-pull).
Collapse
Affiliation(s)
- Breanna Hodgins
- 1Department of Experimental Medicine, McGill University, Montreal, Quebec Canada
| | - Stephane Pillet
- 2Research Institute of McGill University Health Centre, 1001 Boul Decarie, Room EM33248, Montreal, QC H4A 3J1 Canada.,3Medicago Inc., Quebec City, Quebec Canada
| | | | - Brian J Ward
- 2Research Institute of McGill University Health Centre, 1001 Boul Decarie, Room EM33248, Montreal, QC H4A 3J1 Canada.,3Medicago Inc., Quebec City, Quebec Canada
| |
Collapse
|
2
|
Alipour S, Mahdavi A, Abdoli A. The effects of CpG-ODNs and Chitosan adjuvants on the elicitation of immune responses induced by the HIV-1-Tat-based candidate vaccines in mice. Pathog Dis 2017; 75:2975569. [PMID: 28175274 DOI: 10.1093/femspd/ftx013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 02/06/2017] [Indexed: 12/22/2022] Open
Abstract
HIV1-Tat-based vaccines could elicit broad, durable and neutralizing immune responses and are considered as potential AIDS vaccines. The present study aims to formulate CpG-ODNs adjuvant and Chitosan with Tat protein to enhance the immunogenicity of HIV-1-Tat-based candidate vaccines and to investigate their efficacies in mice. To this end, we added CpG-ODNs, Chitosan and Alum as adjuvants to the Tat-based candidate vaccine formulations. Then, we compared frequency and magnitude of both humoral and cellular immune responses from mice immunized with the adjuvant-formulated Tat candidate vaccines against those obtained from mice immunized with recombinant Tat protein alone. Mice were subcutaneously immunized three times at 2-week intervals with the candidate vaccines. Measurements of anti-Tat immune responses showed that all vaccinated groups had a good immunity compared to the control groups and developed high levels of both humoral and cellular responses. However, immunized mice with CpG-ODNs, and Chitosan-adjuvanted Tat vaccines elicited stronger T-cell responses (both humoral and cellular immunity) compared to the others. These data suggest that co-administration of recombinant Tat protein with CpG-ODNs and Chitosan may serve as a potential formulation for enhancing of the Tat vaccine-induced immunity and might have effects on shaping Th polarization induced by HIV1-Tat protein vaccines.
Collapse
Affiliation(s)
- Samira Alipour
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 45195-1159, Iran
| | - Atiyeh Mahdavi
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 45195-1159, Iran
| | - Asghar Abdoli
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran 1316943551, Iran
| |
Collapse
|
3
|
Halsey NA, Talaat KR, Greenbaum A, Mensah E, Dudley MZ, Proveaux T, Salmon DA. The safety of influenza vaccines in children: An Institute for Vaccine Safety white paper. Vaccine 2015; 33 Suppl 5:F1-F67. [PMID: 26822822 DOI: 10.1016/j.vaccine.2015.10.080] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 10/02/2015] [Accepted: 10/06/2015] [Indexed: 01/19/2023]
Abstract
Most influenza vaccines are generally safe, but influenza vaccines can cause rare serious adverse events. Some adverse events, such as fever and febrile seizures, are more common in children than adults. There can be differences in the safety of vaccines in different populations due to underlying differences in genetic predisposition to the adverse event. Live attenuated vaccines have not been studied adequately in children under 2 years of age to determine the risks of adverse events; more studies are needed to address this and several other priority safety issues with all influenza vaccines in children. All vaccines intended for use in children require safety testing in the target age group, especially in young children. Safety of one influenza vaccine in children should not be extrapolated to assumed safety of all influenza vaccines in children. The low rates of adverse events from influenza vaccines should not be a deterrent to the use of influenza vaccines because of the overwhelming evidence of the burden of disease due to influenza in children.
Collapse
Affiliation(s)
- Neal A Halsey
- Department of International Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States; Institute for Vaccine Safety, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States.
| | - Kawsar R Talaat
- Institute for Vaccine Safety, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States; Center for Immunization Research, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Adena Greenbaum
- Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Eric Mensah
- Department of International Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Matthew Z Dudley
- Department of International Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States; Institute for Vaccine Safety, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Tina Proveaux
- Department of International Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States; Institute for Vaccine Safety, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Daniel A Salmon
- Department of International Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States; Institute for Vaccine Safety, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| |
Collapse
|
4
|
Sheets RL, Zhou T, Knezevic I. Scientific and regulatory challenges in evaluating clinical trial protocols for HIV-1/AIDS vaccines - A review from a regulatory perspective. Biologicals 2015; 44:90-110. [PMID: 26732973 DOI: 10.1016/j.biologicals.2015.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 11/20/2015] [Accepted: 11/21/2015] [Indexed: 11/29/2022] Open
Abstract
Clinical development of prophylactic HIV/AIDS vaccines presents many scientific challenges that result in challenges for regulators reviewing clinical trial applications (CTAs). The World Health Organization (WHO) has the responsibility to provide technical support to these regulators. The search for an HIV/AIDS vaccine will only succeed through well-designed, -conducted and -controlled human efficacy studies reviewed and approved by regulators in countries worldwide, particularly in countries where the epidemic has hit hardest, such as in sub-Saharan Africa and Asia. This review summarizes the current candidates in development and focuses on challenges regulators face when reviewing CTAs, such as the evolving landscape of "standard of prevention," trials in adolescents, adaptive trial designs, correlates of protection and their analysis, and access to successful vaccines. There are many unknowns in the field of HIV/AIDS vaccine development and often, there is not a clear right or wrong approach because of the scientific challenges described in this review. Consequently, regulators should not feel that decisions need be made in isolation, when there are many available international collaborative efforts and opportunities to seek expert advice. The WHO provides many such opportunities and support to regulators across the globe.
Collapse
Affiliation(s)
| | - TieQun Zhou
- Technologies Standards and Norms Team, Regulation of Medicines and Other Health Technologies, Department of Essential Medicines and Health Products, Health Systems and Innovation, World Health Organization, Avenue Appia 20, 1211 Geneva 27, Switzerland.
| | - Ivana Knezevic
- Technologies Standards and Norms Team, Regulation of Medicines and Other Health Technologies, Department of Essential Medicines and Health Products, Health Systems and Innovation, World Health Organization, Avenue Appia 20, 1211 Geneva 27, Switzerland.
| |
Collapse
|
5
|
Lorenzen E, Follmann F, Jungersen G, Agerholm JS. A review of the human vs. porcine female genital tract and associated immune system in the perspective of using minipigs as a model of human genital Chlamydia infection. Vet Res 2015; 46:116. [PMID: 26411309 PMCID: PMC4586017 DOI: 10.1186/s13567-015-0241-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 08/11/2015] [Indexed: 03/16/2023] Open
Abstract
Sexually transmitted diseases constitute major health issues and their prevention and treatment continue to challenge the health care systems worldwide. Animal models are essential for a deeper understanding of the diseases and the development of safe and protective vaccines. Currently a good predictive non-rodent model is needed for the study of genital chlamydia in women. The pig has become an increasingly popular model for human diseases due to its close similarities to humans. The aim of this review is to compare the porcine and human female genital tract and associated immune system in the perspective of genital Chlamydia infection. The comparison of women and sows has shown that despite some gross anatomical differences, the structures and proportion of layers undergoing cyclic alterations are very similar. Reproductive hormonal cycles are closely related, only showing a slight difference in cycle length and source of luteolysing hormone. The epithelium and functional layers of the endometrium show similar cyclic changes. The immune system in pigs is very similar to that of humans, even though pigs have a higher percentage of CD4(+)/CD8(+) double positive T cells. The genital immune system is also very similar in terms of the cyclic fluctuations in the mucosal antibody levels, but differs slightly regarding immune cell infiltration in the genital mucosa - predominantly due to the influx of neutrophils in the porcine endometrium during estrus. The vaginal flora in Göttingen Minipigs is not dominated by lactobacilli as in humans. The vaginal pH is around 7 in Göttingen Minipigs, compared to the more acidic vaginal pH around 3.5-5 in women. This review reveals important similarities between the human and porcine female reproductive tracts and proposes the pig as an advantageous supplementary model of human genital Chlamydia infection.
Collapse
Affiliation(s)
- Emma Lorenzen
- Section for Veterinary Reproduction and Obstetrics, Department of Large Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
- Chlamydia Vaccine Research, Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark.
| | - Frank Follmann
- Chlamydia Vaccine Research, Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark.
| | - Gregers Jungersen
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Copenhagen, Denmark.
| | - Jørgen S Agerholm
- Section for Veterinary Reproduction and Obstetrics, Department of Large Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
6
|
McClarty LM, Lorway RR, Ramanaik S, Wylie J, Becker ML. Factors influencing frontline health service providers' likelihood to recommend a future, preventive HIV vaccine to key populations in Karnataka, south India. Vaccine 2014; 33:656-63. [PMID: 25528520 DOI: 10.1016/j.vaccine.2014.12.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 12/03/2014] [Accepted: 12/05/2014] [Indexed: 11/29/2022]
Abstract
The HIV epidemic in the south Indian state of Karnataka disproportionately burdens key populations of men who have sex with men and female sex workers. Despite having successfully reduced HIV incidence among certain key populations through the use of targeted intervention, India's HIV epidemic remains one of its greatest public health issues. The best long-term strategy for managing the global HIV epidemic might involve a preventive vaccine; however, vaccine availability cannot guarantee its accessibility or acceptability. Vaccine recommendations from frontline health service providers have previously been identified as useful strategies to enhance vaccine uptake among target groups. This study used structured interviews to explore frontline health service providers' self-identified likelihood to recommend a future, preventive HIV vaccine to key populations in Karnataka. A modified social ecological model was then used to categorise factors that might prevent health service providers from recommending an HIV vaccine. Overall, 83% of health service providers reported that they would be very likely to recommend an HIV vaccine to men who have sex with men and female sex workers, while less than one-third of participants identified one or more barrier to vaccine recommendation. Intrapersonal, interpersonal, and structural/political factors were most commonly reported to act as potential barriers to future HIV vaccine recommendation among health service providers in Karnataka. This study adds to the limited body of literature focussing on future HIV vaccine acceptability in low- and middle-income countries and highlights some of the several complexities surrounding vaccine acceptability and uptake among key populations in Karnataka.
Collapse
Affiliation(s)
- Leigh M McClarty
- Centre for Global Public Health, Department of Community Health Sciences, University of Manitoba, R070 Medical Rehabilitation Building, 771 McDermot Avenue, Winnipeg, Manitoba R3E 0T6, Canada.
| | - Robert R Lorway
- Centre for Global Public Health, Department of Community Health Sciences, University of Manitoba, R070 Medical Rehabilitation Building, 771 McDermot Avenue, Winnipeg, Manitoba R3E 0T6, Canada.
| | - Satyanarayana Ramanaik
- Karnataka Health Promotion Trust, IT Park, #1-4, 5th Floor, Rajajinagar Industrial Area, Rajajinagar, Bangalore, 560044, India.
| | - John Wylie
- Department of Medical Microbiology, University of Manitoba, 543 Basic Medical Sciences Building, 745 Bannatyne Avenue, Winnipeg, Manitoba R3E 0J9, Canada; Department of Community Health Sciences, University of Manitoba, S113 Medical Services Building, 750 Bannatyne Avenue, Winnipeg, Manitoba R3E 0W3, Canada.
| | - Marissa L Becker
- Centre for Global Public Health, Department of Community Health Sciences, University of Manitoba, R070 Medical Rehabilitation Building, 771 McDermot Avenue, Winnipeg, Manitoba R3E 0T6, Canada; Department of Community Health Sciences, University of Manitoba, S113 Medical Services Building, 750 Bannatyne Avenue, Winnipeg, Manitoba R3E 0W3, Canada.
| |
Collapse
|
7
|
Segalés J. Best practice and future challenges for vaccination against porcine circovirus type 2. Expert Rev Vaccines 2014; 14:473-87. [DOI: 10.1586/14760584.2015.983084] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Joaquim Segalés
- Centre de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
- Departament de Sanitat i d’Anatomia Animals, Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| |
Collapse
|
8
|
Abstract
Summary Between July 2005 and January 2006 we evaluated 1248 Paraguayan active duty military volunteers. Participants provided a blood sample for HIV testing and answered an anonymous survey. HIV seroprevalence was 0.4% (5 of 1248) among participants. The median age at first sexual intercourse was 16 years. Only 14.8% of participants reported condom use with every sexual encounter. Military students used condoms the most. Participants older than 45 years, compared with younger participants, had a fourfold (adjusted odds ratio 4.3) increased risk of not using condoms. Men were less likely to use a condom, more likely to practice anal intercourse, and had more sexual partners than women. Officers and non-commissioned officers were identified to have a twofold (as measured by adjusted odds ratio = 2.00 and 2.22, respectively) increased risk of having more than two sexual partners in the last month compared with students. Both officers and non-commissioned officers were twice as likely as students to practice anal intercourse. Despite the high-risk behaviours reported by those surveyed, HIV seroprevalence in active duty personnel was low. Future efforts should emphasize on the correct condom use keeping focus on the high-risk behaviours of groups at risk, and on routinely testing the military personnel for HIV.
Collapse
|
9
|
Abstract
PURPOSE OF REVIEW To briefly describe some of the replication-competent vectors being investigated for development of candidate HIV vaccines focusing primarily on technologies that have advanced to testing in macaques or have entered clinical trials. RECENT FINDINGS Replication-competent viral vectors have advanced to the stage at which decisions can be made regarding the future development of HIV vaccines. The viruses being used as replication-competent vector platforms vary considerably, and their unique attributes make it possible to test multiple vaccine design concepts and also mimic various aspects of an HIV infection. Replication-competent viral vectors encoding simian immunodeficiency virus or HIV proteins can be used to safely immunize macaques, and in some cases, there is evidence of significant vaccine efficacy in challenge protection studies. Several live HIV vaccine vectors are in clinical trials to evaluate immunogenicity, safety, the effect of mucosal delivery, and potential effects of preexisting immunity. SUMMARY A variety of DNA and RNA viruses are being used to develop replication-competent viral vectors for HIV vaccine delivery. Multiple viral vector platforms have proven to be well tolerated and immunogenic with evidence of efficacy in macaques. Some of the more advanced HIV vaccine prototypes based on vesicular stomatitis virus, vaccinia virus, measles virus, and Sendai virus are in clinical trials.
Collapse
|
10
|
Abstract
PURPOSE OF REVIEW Early studies have cast doubt on the utility of animal models for predicting success or failure of HIV-prevention strategies, but results of multiple human phase 3 microbicide trials, and interrogations into the discrepancies between human and animal model trials, indicate that animal models were, and are, predictive of safety and efficacy of microbicide candidates. RECENT FINDINGS Recent studies have shown that topically applied vaginal gels, and oral prophylaxis using single or combination antiretrovirals are indeed effective in preventing sexual HIV transmission in humans, and all of these successes were predicted in animal models. Further, prior discrepancies between animal and human results are finally being deciphered as inadequacies in study design in the model, or quite often, noncompliance in human trials, the latter being increasingly recognized as a major problem in human microbicide trials. SUMMARY Successful microbicide studies in humans have validated results in animal models, and several ongoing studies are further investigating questions of tissue distribution, duration of efficacy, and continued safety with repeated application of these, and other promising microbicide candidates in both murine and nonhuman primate models. Now that we finally have positive correlations with prevention strategies and protection from HIV transmission, we can retrospectively validate animal models for their ability to predict these results, and more importantly, prospectively use these models to select and advance even safer, more effective, and importantly, more durable microbicide candidates into human trials.
Collapse
|
11
|
Bon I, Lembo D, Rusnati M, Clò A, Morini S, Miserocchi A, Bugatti A, Grigolon S, Musumeci G, Landolfo S, Re MC, Gibellini D. Peptide-derivatized SB105-A10 dendrimer inhibits the infectivity of R5 and X4 HIV-1 strains in primary PBMCs and cervicovaginal histocultures. PLoS One 2013; 8:e76482. [PMID: 24116111 PMCID: PMC3792046 DOI: 10.1371/journal.pone.0076482] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 08/30/2013] [Indexed: 11/19/2022] Open
Abstract
Peptide dendrimers are a class of molecules that exhibit a large array of biological effects including antiviral activity. In this report, we analyzed the antiviral activity of the peptide-derivatized SB105-A10 dendrimer, which is a tetra-branched dendrimer synthetized on a lysine core, in activated peripheral blood mononuclear cells (PBMCs) that were challenged with reference and wild-type human immunodeficiency virus type 1 (HIV-1) strains. SB105-A10 inhibited infections by HIV-1 X4 and R5 strains, interfering with the early phases of the viral replication cycle. SB105-A10 targets heparan sulfate proteoglycans (HSPGs) and, importantly, the surface plasmon resonance (SPR) assay revealed that SB105-A10 strongly binds gp41 and gp120, most likely preventing HIV-1 attachment/entry through multiple mechanisms. Interestingly, the antiviral activity of SB105-A10 was also detectable in an organ-like structure of human cervicovaginal tissue, in which SB105-A10 inhibited the HIV-1ada R5 strain infection without altering the tissue viability. These results demonstrated the strong antiviral activity of SB105-A10 and suggest a potential microbicide use of this dendrimer to prevent the heterosexual transmission of HIV-1.
Collapse
Affiliation(s)
- Isabella Bon
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Microbiology Section, University of Bologna, Bologna, Italy
| | - David Lembo
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino, Italy
| | - Marco Rusnati
- Department of Biomedical Sciences and Biotechnology, University of Brescia, Brescia, Italy
| | - Alberto Clò
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Microbiology Section, University of Bologna, Bologna, Italy
| | - Silvia Morini
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Microbiology Section, University of Bologna, Bologna, Italy
| | - Anna Miserocchi
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Microbiology Section, University of Bologna, Bologna, Italy
| | - Antonella Bugatti
- Department of Biomedical Sciences and Biotechnology, University of Brescia, Brescia, Italy
| | | | - Giuseppina Musumeci
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Microbiology Section, University of Bologna, Bologna, Italy
| | - Santo Landolfo
- Department of Public Health and Microbiology, University of Torino, Torino, Italy
| | - Maria Carla Re
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Microbiology Section, University of Bologna, Bologna, Italy
- Interuniversity Consortium, National Institute Biostructure and Biosystems (INBB) Roma, Italy
| | - Davide Gibellini
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Microbiology Section, University of Bologna, Bologna, Italy
| |
Collapse
|
12
|
Jackson A, Kløverpris HN, Boffito M, Handley A, Atkins M, Hayes P, Gilmour J, Riddel L, Chen F, Bailey-Tippets M, Walker B, Ackland J, Sullivan M, Goulder P. A randomised, placebo-controlled, first-in-human study of a novel clade C therapeutic peptide vaccine administered ex vivo to autologous white blood cells in HIV infected individuals. PLoS One 2013; 8:e73765. [PMID: 24069230 PMCID: PMC3775760 DOI: 10.1371/journal.pone.0073765] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 07/17/2013] [Indexed: 11/21/2022] Open
Abstract
Background Preclinical studies of overlapping 15mer peptides, spanning SIV, SHIV or HIV, pulsed on autologous PBMC ex vivo have demonstrated high level, virus-specific T cell responses and viral suppression in non-human primates (NHP). Opal-HIV-Gag(c) consists of 120 synthetic 15mer peptides spanning Clade C, consensus Gag, manufactured to current good manufacturing practice; having been evaluated in a good laboratory practice toxicology study in Macaca mulatta. We evaluated the safety and preliminary immunogenicity of such peptides administered intravenously after short-duration ex vivo incubation, to HIV-positive adults on suppressive antiretroviral therapy. Methods and Findings A first-in-human, placebo-controlled, double-blind, dose escalation study was conducted. Twenty-three patients with virus suppressed by antiretroviral therapy were enrolled in four groups 12 mg (n = 6), 24 mg (n = 6), 48 mg (n = 2) or matching placebo (n = 8). Treatment was administered intravenously after bedside enrichment of 120 mL whole blood for white cells using a closed system (Sepax S-100 device), with ex vivo peptide admixture (or diluent alone) and 37°C incubation for one hour prior to reinfusion. Patients received 4 administrations at monthly intervals followed by a 12-week observation post-treatment. Opal-HIV-Gag(c) was reasonably tolerated at doses of 12 and 24 mg. There was an increased incidence of temporally associated pyrexia, chills, and transient/self-limiting lymphopenia in Opal-HIV-Gag(c) recipients compared to placebo. The study was terminated early, after two patients were recruited to the 48 mg cohort; a serious adverse event of hypotension, tachycardia secondary to diarrhoea occurred following a single product administration. An infectious cause for the event could not be identified, leaving the possibility of immunologically mediated product reaction. Conclusions A serious, potentially life-threatening event of hypotension led to early, precautionary termination of the study. In the absence of a clearly defined mechanism or ability to predict such occurrence, further development of Opal-HIV-Gag(c) will not be undertaken in the current form. Registration ClinicalTrials.gov NCT01123915; EudraCT: 2008-005142-23
Collapse
Affiliation(s)
- Akil Jackson
- St Stephen’s AIDS Trust, Chelsea and Westminster Hospital, London, United Kingdom
- * E-mail:
| | - Henrik N. Kløverpris
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- Department of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- KwaZulu-Natal Research Institute for Tuberculosis and HIV, K-RITH, University of Kwazulu-Natal, Durban, South Africa
| | - Marta Boffito
- St Stephen’s AIDS Trust, Chelsea and Westminster Hospital, London, United Kingdom
| | | | | | | | | | - Lynn Riddel
- Department of Genitourinary Medicine, Northampton General Hospital, Northampton, United Kingdom
| | - Fabian Chen
- Department of Sexual Health, Royal Berkshire Hospital, Reading, United Kingdom
| | | | - Bruce Walker
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Boston, Massachusetts, United States of America
| | - Jim Ackland
- Global Biosolutions, Craigieburn, Victoria, Australia
| | - Mark Sullivan
- Medicines Development, Melbourne, Victoria, Australia
| | - Philip Goulder
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Boston, Massachusetts, United States of America
| |
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW One of the major obstacles in fully understanding HIV transmission comes from the impracticality of studying transmission in humans. Because of this encumbrance, the early phases of HIV transmission and systemic dissemination are poorly understood. In order to fully comprehend these critical steps in HIV infection, animal models must be devised to accurately reflect HIV's mode of action. This review seeks to highlight the essential nature of modelling HIV transmission in nonhuman primates (NHPs). RECENT FINDINGS Recently, it was discovered that HIV infection is established in newly infected recipients by a single or few transmitted/founder variants. This has reshaped how animal modelling is conducted with researchers currently recapitulating a physiologically relevant, low-titre infection. Pertinent animal models have been established for the most common routes of infection, including rectal, vaginal and penile transmission; models for intravenous and oral transmission are still in developmental stages. SUMMARY These limited dose models now accurately reflect HIV transmission in humans and provide a realistic experimental platform for vaccine development and other intervention strategies that can be used to inform vaccine development in humans. Using information obtained in NHP and human trials, it is conceivable to envision effective prevention modalities in the near future.
Collapse
Affiliation(s)
- Christine M. Fennessey
- AIDS and Cancer Virus Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Brandon F. Keele
- AIDS and Cancer Virus Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| |
Collapse
|
14
|
Topological analysis of HIV-1 glycoproteins expressed in situ on virus surfaces reveals tighter packing but greater conformational flexibility than for soluble gp120. J Virol 2013; 87:9233-49. [PMID: 23740975 DOI: 10.1128/jvi.01145-13] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In natural infection, antibodies interact with HIV-1 primarily through nonfunctional forms of envelope glycoproteins (Env), including uncleaved (UNC) gp160 and gp41 stumps. These antigens are important to fully characterize, as they may be decoys that promote nonneutralizing responses and may also be targets for nonneutralizing effector responses. In this study, we compared the antigenic properties of Env expressed in situ on pseudovirion virus-like particle (VLP) surfaces and soluble gp120 using harmonized enzyme-linked immunosorbent assays (ELISAs) and a panel of 51 monoclonal antibodies (MAbs). Only 32 of 46 soluble gp120-reactive MAbs recognized the primary UNC gp160 antigen of VLPs. Indeed, many epitopes were poorly exposed (C1, V2, C1-C4, C4, C4-V3, CD4 induced [CD4i], and PGT group 3) or obscured (C2, C5, and C1-C5) on VLPs. In further studies, VLP Env exhibited an increased degree of inter-MAb competition, the epicenter of which was the base of the V3 loop, where PGT, 2G12, V3, and CD4 binding site specificities competed. UNC gp160 also underwent more drastic soluble CD4 (sCD4)-induced conformational changes than soluble gp120, exposing CD4i, C1-C4, and V2 epitopes. A greater propensity of UNC gp160 to undergo conformational changes was also suggested by the induction of CD4i MAb binding to VLPs by a V3 MAb as well as by soluble CD4. The same effect was not observed for soluble gp120. Taken together, our data suggest that membrane-expressed UNC gp160 exists in a less "triggered" conformational state than soluble gp120 and that MAb binding to UNC gp160 tends to have greater conformational consequences.
Collapse
|
15
|
Kunwar P, Hawkins N, Dinges WL, Liu Y, Gabriel EE, Swan DA, Stevens CE, Maenza J, Collier AC, Mullins JI, Hertz T, Yu X, Horton H. Superior control of HIV-1 replication by CD8+ T cells targeting conserved epitopes: implications for HIV vaccine design. PLoS One 2013; 8:e64405. [PMID: 23741326 PMCID: PMC3669284 DOI: 10.1371/journal.pone.0064405] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 04/12/2013] [Indexed: 12/21/2022] Open
Abstract
A successful HIV vaccine will likely induce both humoral and cell-mediated immunity, however, the enormous diversity of HIV has hampered the development of a vaccine that effectively elicits both arms of the adaptive immune response. To tackle the problem of viral diversity, T cell-based vaccine approaches have focused on two main strategies (i) increasing the breadth of vaccine-induced responses or (ii) increasing vaccine-induced responses targeting only conserved regions of the virus. The relative extent to which set-point viremia is impacted by epitope-conservation of CD8+ T cell responses elicited during early HIV-infection is unknown but has important implications for vaccine design. To address this question, we comprehensively mapped HIV-1 CD8+ T cell epitope-specificities in 23 ART-naïve individuals during early infection and computed their conservation score (CS) by three different methods (prevalence, entropy and conseq) on clade-B and group-M sequence alignments. The majority of CD8+ T cell responses were directed against variable epitopes (p<0.01). Interestingly, increasing breadth of CD8+ T cell responses specifically recognizing conserved epitopes was associated with lower set-point viremia (r = - 0.65, p = 0.009). Moreover, subjects possessing CD8+ T cells recognizing at least one conserved epitope had 1.4 log10 lower set-point viremia compared to those recognizing only variable epitopes (p = 0.021). The association between viral control and the breadth of conserved CD8+ T cell responses may be influenced by the method of CS definition and sequences used to determine conservation levels. Strikingly, targeting variable versus conserved epitopes was independent of HLA type (p = 0.215). The associations with viral control were independent of functional avidity of CD8+ T cell responses elicited during early infection. Taken together, these data suggest that the next-generation of T-cell based HIV-1 vaccines should focus on strategies that can elicit CD8+ T cell responses to multiple conserved epitopes of HIV-1.
Collapse
Affiliation(s)
- Pratima Kunwar
- Viral Vaccine Program, Seattle Biomedical Research Institute, Seattle, Washington, United States of America
- Department of Global Health, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Natalie Hawkins
- Statistical Center for HIV Research and Prevention, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Warren L. Dinges
- Viral Vaccine Program, Seattle Biomedical Research Institute, Seattle, Washington, United States of America
- Polyclinic Infectious Disease, Seattle, Washington, United States of America
| | - Yi Liu
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Erin E. Gabriel
- Statistical Center for HIV Research and Prevention, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - David A. Swan
- Statistical Center for HIV Research and Prevention, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Claire E. Stevens
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Janine Maenza
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Ann C. Collier
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - James I. Mullins
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
- Department of Laboratory Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Tomer Hertz
- Statistical Center for HIV Research and Prevention, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Xuesong Yu
- Statistical Center for HIV Research and Prevention, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Helen Horton
- Viral Vaccine Program, Seattle Biomedical Research Institute, Seattle, Washington, United States of America
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- Department of Global Health, University of Washington School of Medicine, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
16
|
|
17
|
Naim HY. Applications and challenges of multivalent recombinant vaccines. Hum Vaccin Immunother 2012; 9:457-61. [PMID: 23249651 DOI: 10.4161/hv.23220] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The exceptional discoveries of antigen/gene delivery systems have allowed the development of novel prophylactic and therapeutic vaccine candidates. The vaccine candidates employ various antigen-delivery systems, particularly recombinant viral vectors. Recombinant viral vectors are experimental vaccines similar to DNA vaccines, but they use attenuated viruses or bacterium as a carrier "vector" to introduce microbial DNA to cells of the body. They closely mimic a natural infection and therefore can efficiently stimulate the immune system. Although such recombinant vectors may face extensive preclinical testing and will possibly have to meet stringent regulatory requirements, some of these vectors (e.g. measles virus vectors) may benefit from the profound industrial and clinical experience of the parent vaccine. Most notably, novel vaccines based on live attenuated viruses combine the induction of broad, strong and persistent immune responses with acceptable safety profiles. We assess certain technologies in light of their use against human immunodeficiency virus (HIV).
Collapse
Affiliation(s)
- Hussein Y Naim
- Institute of Molecular Biology; University of Zürich-Irchel; Zürich, Switzerland; Current affiliation: Consultant; Life Sciences and Vaccines; Bern, Switzerland
| |
Collapse
|
18
|
Schiller JT, Lowy DR. Understanding and learning from the success of prophylactic human papillomavirus vaccines. Nat Rev Microbiol 2012; 10:681-92. [PMID: 22961341 PMCID: PMC6309166 DOI: 10.1038/nrmicro2872] [Citation(s) in RCA: 160] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
An estimated 5% of human cancers are caused by human papillomavirus (HPV) infections, and most of these cancers are of the cervix. Two prophylactic HPV vaccines that target the two most oncogenic virus types, HPV16 and HPV18, are now commercially available. In controlled clinical trials, the vaccines proved to be effective at preventing incident anogenital infection and the associated neoplastic disease that is induced by these virus types. Here, we highlight the specific aspects of HPV biology and vaccine composition that are likely to contribute to the efficacy of these vaccines, and we discuss how these particular features might or might not be relevant for the development of effective vaccines against other sexually transmitted viruses such as HIV and herpes simplex virus (HSV).
Collapse
Affiliation(s)
- John T Schiller
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | |
Collapse
|
19
|
HIV reservoirs and immune surveillance evasion cause the failure of structured treatment interruptions: a computational study. PLoS One 2012; 7:e36108. [PMID: 22558348 PMCID: PMC3338637 DOI: 10.1371/journal.pone.0036108] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 03/30/2012] [Indexed: 11/19/2022] Open
Abstract
Continuous antiretroviral therapy is currently the most effective way to treat HIV infection. Unstructured interruptions are quite common due to side effects and toxicity, among others, and cannot be prevented. Several attempts to structure these interruptions failed due to an increased morbidity compared to continuous treatment. The cause of this failure is poorly understood and often attributed to drug resistance. Here we show that structured treatment interruptions would fail regardless of the emergence of drug resistance. Our computational model of the HIV infection dynamics in lymphoid tissue inside lymph nodes, demonstrates that HIV reservoirs and evasion from immune surveillance themselves are sufficient to cause the failure of structured interruptions. We validate our model with data from a clinical trial and show that it is possible to optimize the schedule of interruptions to perform as well as the continuous treatment in the absence of drug resistance. Our methodology enables studying the problem of treatment optimization without having impact on human beings. We anticipate that it is feasible to steer new clinical trials using computational models.
Collapse
|