201
|
Abstract
Progress in vector design and an increased knowledge of mechanisms underlying tumor-induced immune suppression have led to a new and promising generation of Adenovirus (Ad)-based immunotherapies, which are discussed in this review. As vaccine vehicles Ad vectors (AdVs) have been clinically evaluated and proven safe, but a major limitation of the commonly used Ad5 serotype is neutralization by preexistent or rapidly induced immune responses. Genetic modifications in the Ad capsid can reduce intrinsic immunogenicity and facilitate escape from antibody-mediated neutralization. Further modification of the Ad hexon and fiber allows for liver and scavenger detargeting and selective targeting of, for example, dendritic cells. These next-generation Ad vaccines with enhanced efficacy are now becoming available for testing as tumor vaccines. In addition, AdVs encoding immune-modulating products may be used to convert the tumor microenvironment from immune-suppressive and proinvasive to proinflammatory, thus facilitating cell-mediated effector functions that can keep tumor growth and invasion in check. Oncolytic AdVs, that selectively replicate in tumor cells and induce an immunogenic form of cell death, can also be armed with immune-activating transgenes to amplify primed antitumor immune responses. These novel immunotherapy strategies, employing highly efficacious AdVs in optimized configurations, show great promise and warrant clinical exploration.
Collapse
|
202
|
Abstract
Human adenoviral vectors are being developed for use in candidate vaccines for HIV-1 and other pathogens. However, this approach suffered a setback when an HIV-1 vaccine using an adenovirus type 5 (Ad5) vector failed to reduce, and might even have increased, the rate of HIV infection in men who were uncircumcised and who had preexisting antibodies specific for Ad5. This increased interest in the evaluation of serologically distinct adenoviral vectors. In this issue of the JCI, Frahm and coworkers report evidence that preexisting cellular immune responses directed toward Ad5 reduce the immunogenicity of antigens expressed in Ad5-vectored vaccines and have cross-reacting potential with non-Ad5 adenoviral vectors. The implications of this observation need to be carefully evaluated in future clinical trials of all serotypes of adenovirus-vectored vaccines.
Collapse
Affiliation(s)
- Nelson L Michael
- US Military HIV Research Program, Walter Reed Army Institute of Research, Bethesda, Maryland 20817, USA.
| |
Collapse
|
203
|
Choi JH, Schafer SC, Zhang L, Kobinger GP, Juelich T, Freiberg AN, Croyle MA. A single sublingual dose of an adenovirus-based vaccine protects against lethal Ebola challenge in mice and guinea pigs. Mol Pharm 2011; 9:156-67. [PMID: 22149096 DOI: 10.1021/mp200392g] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Sublingual (SL) delivery, a noninvasive immunization method that bypasses the intestinal tract for direct entry into the circulation, was evaluated with an adenovirus (Ad5)-based vaccine for Ebola. Mice and guinea pigs were immunized via the intramuscular (IM), nasal (IN), oral (PO) and SL routes. SL immunization elicited strong transgene expression in and attracted CD11c(+) antigen presenting cells to the mucosa. A SL dose of 1 × 10⁸ infectious particles induced Ebola Zaire glycoprotein (ZGP)-specific IFN-γ⁺ T cells in spleen, bronchoalveolar lavage, mesenteric lymph nodes and submandibular lymph nodes (SMLN) of naive mice in a manner similar to the same dose given IN. Ex vivo CFSE and in vivo cytotoxic T lymphocyte (CTL) assays confirmed that SL immunization elicits a notable population of effector memory CD8+ T cells and strong CTL responses in spleen and SMLN. SL immunization induced significant ZGP-specific Th1 and Th2 type responses unaffected by pre-existing immunity (PEI) that protected mice and guinea pigs from lethal challenge. SL delivery protected more mice with PEI to Ad5 than IM injection. SL immunization also reduced systemic anti-Ad5 T and B cell responses in naive mice and those with PEI, suggesting that secondary immunizations could be highly effective for both populations.
Collapse
Affiliation(s)
- Jin Huk Choi
- Division of Pharmaceutics, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712, United States
| | | | | | | | | | | | | |
Collapse
|
204
|
Lam E, Ramke M, Groos S, Warnecke G, Heim A. A differentiated porcine bronchial epithelial cell culture model for studying human adenovirus tropism and virulence. J Virol Methods 2011; 178:117-23. [PMID: 21907242 DOI: 10.1016/j.jviromet.2011.08.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 08/18/2011] [Accepted: 08/22/2011] [Indexed: 10/17/2022]
Abstract
The species specificity of human adenoviruses (HAdV) almost precludes studying virulence and tropism in animal models, e.g. rodent models, or derived tissue and cell culture models. However, replication of HAdV type 5 (HAdV-C5) has been shown after intravenous injection in swine. In order to study adenovirus replication in airway tissue propagation of bronchial epithelial cells from porcine lungs was established. These primary cells proved to be fully permissive for HAdV-C5 infection in submerged culture, demonstrating efficient HAdV genome replication, infectious viral particle release (1.07×10(8) TCID(50)/ml±6.63×10(7)) and development of cytopathic effect (CPE). Differentiation of porcine bronchial epithelial cells was achieved at the air-liquid interface on collagen I coated 0.4μm polyester membranes. Morphology, expression of tubulin and occludin, the development of tight-junctions and cilia were similar to human bronchial epithelial cells. Infection with HAdV-C5 from the basolateral side resulted in release of infectious virus progeny (2.05×10(7) TCID(50)/ml±2.39×10(7)) to the apical surface as described recently in human bronchial epithelial cells, although complete CPE was not observed. Differentiated porcine bronchial epithelial cells hold promise as a novel method for studying the virulence and pathophysiology of pneumonia associated HAdV types.
Collapse
Affiliation(s)
- E Lam
- Institute of Virology, Hannover Medical School, Carl-Neubergstr. 1, 30625 Hannover, Germany.
| | | | | | | | | |
Collapse
|
205
|
Gabitzsch ES, Xu Y, Balint JP, Balcaitis S, Sanders-Beer B, Jones FR. Induction and comparison of SIV immunity in Ad5 naïve and Ad5 immune non-human primates using an Ad5 [E1-, E2b-] based vaccine. Vaccine 2011; 29:8101-7. [PMID: 21864618 DOI: 10.1016/j.vaccine.2011.08.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Revised: 06/16/2011] [Accepted: 08/06/2011] [Indexed: 01/06/2023]
Abstract
The effectiveness of recombinant Adenovirus serotype 5 (Ad5) vectors to induce immune responses against targeted antigens has been limited by the presence of pre-existing or Ad5 vaccine induced anti-vector immunity. The Ad5 [E1-, E2b-] platform, a recombinant Ad5 with additional deletions, has been previously reported by us to induce immune responses in the presence of Ad5 immunity. In an Ad5 immune non-human primate (NHP) model, an Ad5 [E1-, E2b-] construct expressing HIV-1 Gag induced immune responses in the presence of pre-existing Ad5 immunity. In the present study we expand on these prior observations by comparing the cell mediated immune (CMI) responses induced by Ad5 [E1-, E2b-]-SIV-gag/nef in Ad5 naïve and Ad5 immune NHP. Additionally, NHP were immunized with an Ad5 [E1-, E2b-]-HIV-pol construct following two homologous administrations of Ad5 [E1-, E2b-]-SIV-gag/nef to determine if an immune response could be induced against a third antigen in the presence of vaccine induced Ad5 immunity. Positive CMI responses, as assessed by interferon-gamma (IFN-γ) secreting lymphocytes, were induced against all three antigens. These CMI responses increased over a course of multiple immunizations and the response profiles observed in Ad5 naïve and Ad5 immune NHP were similar. No influence of the major histocompatibility complex on CMI responses was observed. These data indicate that the new Ad5 [E1-, E2b-] platform based vaccine could be used for homologous vaccination regimes to induce robust CMI responses in the presence of Ad5 vector immunity.
Collapse
|
206
|
Churchyard GJ, Morgan C, Adams E, Hural J, Graham BS, Moodie Z, Grove D, Gray G, Bekker LG, McElrath MJ, Tomaras GD, Goepfert P, Kalams S, Baden LR, Lally M, Dolin R, Blattner W, Kalichman A, Figueroa JP, Pape J, Schechter M, Defawe O, De Rosa SC, Montefiori DC, Nabel GJ, Corey L, Keefer MC. A phase IIA randomized clinical trial of a multiclade HIV-1 DNA prime followed by a multiclade rAd5 HIV-1 vaccine boost in healthy adults (HVTN204). PLoS One 2011; 6:e21225. [PMID: 21857901 PMCID: PMC3152265 DOI: 10.1371/journal.pone.0021225] [Citation(s) in RCA: 121] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Accepted: 05/23/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The safety and immunogenicity of a vaccine regimen consisting of a 6-plasmid HIV-1 DNA prime (envA, envB, envC, gagB, polB, nefB) boosted by a recombinant adenovirus serotype-5 (rAd5) HIV-1 with matching inserts was evaluated in HIV-seronegative participants from South Africa, United States, Latin America and the Caribbean. METHODS 480 participants were evenly randomized to receive either: DNA (4 mg i.m. by Biojector) at 0, 1 and 2 months, followed by rAd5 (10(10) PU i.m. by needle/syringe) at 6 months; or placebo. Participants were monitored for reactogenicity and adverse events throughout the 12-month study. Peak and duration of HIV-specific humoral and cellular immune responses were evaluated after the prime and boost. RESULTS The vaccine was well tolerated and safe. T-cell responses, detected by interferon-γ (IFN-γ) ELISpot to global potential T-cell epitopes (PTEs) were observed in 70.8% (136/192) of vaccine recipients overall, most frequently to Gag (54.7%) and to Env (54.2%). In U.S. vaccine recipients T-cell responses were less frequent in Ad5 sero-positive versus sero-negative vaccine recipients (62.5% versus 85.7% respectively, p = 0.035). The frequency of HIV-specific CD4+ and CD8+ T-cell responses detected by intracellular cytokine staining were similar (41.8% and 47.2% respectively) and most secreted ≥2 cytokines. The vaccine induced a high frequency (83.7%-94.6%) of binding antibody responses to consensus Group M, and Clades A, B and C gp140 Env oligomers. Antibody responses to Gag were elicited in 46% of vaccine recipients. CONCLUSION The vaccine regimen was well-tolerated and induced polyfunctional CD4+ and CD8+ T-cells and multi-clade anti-Env binding antibodies. TRIAL REGISTRATION ClinicalTrials.gov NCT00125970.
Collapse
MESH Headings
- AIDS Vaccines/administration & dosage
- AIDS Vaccines/immunology
- Adenoviridae/genetics
- Adolescent
- Adult
- Anemia/chemically induced
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Cohort Studies
- Enzyme-Linked Immunosorbent Assay
- Female
- HIV-1/genetics
- HIV-1/immunology
- Human Immunodeficiency Virus Proteins/genetics
- Human Immunodeficiency Virus Proteins/immunology
- Humans
- Immunization/adverse effects
- Immunization/methods
- Immunization, Secondary/adverse effects
- Immunization, Secondary/methods
- Immunoglobulin G/blood
- Immunoglobulin G/immunology
- Interferon-gamma/blood
- Interferon-gamma/immunology
- Male
- Middle Aged
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/immunology
- Young Adult
- env Gene Products, Human Immunodeficiency Virus/genetics
- env Gene Products, Human Immunodeficiency Virus/immunology
- gag Gene Products, Human Immunodeficiency Virus/genetics
- gag Gene Products, Human Immunodeficiency Virus/immunology
- nef Gene Products, Human Immunodeficiency Virus/genetics
- nef Gene Products, Human Immunodeficiency Virus/immunology
- pol Gene Products, Human Immunodeficiency Virus/genetics
- pol Gene Products, Human Immunodeficiency Virus/immunology
Collapse
Affiliation(s)
- Gavin J. Churchyard
- Aurum Institute for Health Research, Klerksdorp, South Africa
- Centre for AIDS Programme of Research in South Africa (CAPRISA), University of Kwa-Zulu Natal, Durban, South Africa
| | - Cecilia Morgan
- Statistical Center for HIV/AIDS Research and Prevention, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Elizabeth Adams
- Division of AIDS, National Institute of Allergy and Infectious Diseases (NIAID), National Institute of Health (NIH), Bethesda, Maryland, United States of America
| | - John Hural
- Statistical Center for HIV/AIDS Research and Prevention, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Barney S. Graham
- Vaccine Research Center, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Zoe Moodie
- Statistical Center for HIV/AIDS Research and Prevention, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Doug Grove
- Statistical Center for HIV/AIDS Research and Prevention, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Glenda Gray
- Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Linda-Gail Bekker
- Desmond Tutu HIV Foundation, University of Cape Town, Cape Town, South Africa
| | - M. Juliana McElrath
- Statistical Center for HIV/AIDS Research and Prevention, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Georgia D. Tomaras
- Duke University Medical Center, Durham, North Carolina, United States of America
| | - Paul Goepfert
- University of Alabama, Birmingham, Alabama, United States of America
| | - Spyros Kalams
- Vanderbilt University, Nashville, Tennessee, United States of America
| | - Lindsey R. Baden
- Harvard-Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Michelle Lally
- Alpert Medical School of Brown University and Miriam Hospital, Providence, Rhode Island, United States of America
| | - Raphael Dolin
- Harvard Medical School- Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - William Blattner
- University of Maryland, College Park, Maryland, United States of America
| | - Artur Kalichman
- Centro de Referencia e Treinamento em DST/AIDS, Coordenacao dos Institutos de Pesquisa, San Paulo, Brazil
| | | | - Jean Pape
- Cornell-GHESKIO, Institut National de Laboratoire et de Recherches, Port au Prince, Haiti
| | - Mauro Schechter
- Projeto Praça Onze, Hospital Escola São Francisco de Assis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Olivier Defawe
- Statistical Center for HIV/AIDS Research and Prevention, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Stephen C. De Rosa
- Statistical Center for HIV/AIDS Research and Prevention, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - David C. Montefiori
- Duke Human Vaccine Institute, School of Medicine, Duke University, Durham, North Carolina, United States of America
| | - Gary J. Nabel
- Vaccine Research Center, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Lawrence Corey
- Statistical Center for HIV/AIDS Research and Prevention, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Michael C. Keefer
- Department of Medicine, University of Rochester School of Medicine & Dentistry, Rochester, New York, United States of America
| | | |
Collapse
|
207
|
Robb M. HIV vaccine development: a new beginning. Expert Rev Vaccines 2011; 10:925-7. [DOI: 10.1586/erv.11.75] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
208
|
International seroepidemiology of adenovirus serotypes 5, 26, 35, and 48 in pediatric and adult populations. Vaccine 2011; 29:5203-9. [PMID: 21619905 DOI: 10.1016/j.vaccine.2011.05.025] [Citation(s) in RCA: 226] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2011] [Revised: 05/02/2011] [Accepted: 05/11/2011] [Indexed: 11/22/2022]
Abstract
Recombinant adenovirus serotype 5 (rAd5) vaccine vectors for HIV-1 and other pathogens have been shown to be limited by high titers of Ad5 neutralizing antibodies (NAbs) in the developing world. Alternative serotype rAd vectors have therefore been constructed. Here we report Ad5, Ad26, Ad35, and Ad48 NAb titers in 4381 individuals from North America, South America, sub-Saharan Africa, and Southeast Asia. As expected, Ad5 NAb titers were both frequent and high magnitude in sub-Saharan Africa and Southeast Asia. In contrast, Ad35 NAb titers proved infrequent and low in all regions studied, and Ad48 NAbs were rare in all regions except East Africa. Ad26 NAbs were moderately common in adults in sub-Saharan Africa and Southeast Asia, but Ad26 NAb titers proved markedly lower than Ad5 NAb titers in all regions, and these relatively low Ad26 NAb titers did not detectably suppress the immunogenicity of 4×10(10)vp of a rAd26-Gag/Pol/Env/Nef vaccine in rhesus monkeys. These data inform the clinical development of alternative serotype rAd vaccine vectors in the developing world.
Collapse
|
209
|
Vasan S, Hurley A, Schlesinger SJ, Hannaman D, Gardiner DF, Dugin DP, Boente-Carrera M, Vittorino R, Caskey M, Andersen J, Huang Y, Cox JH, Tarragona-Fiol T, Gill DK, Cheeseman H, Clark L, Dally L, Smith C, Schmidt C, Park HH, Kopycinski JT, Gilmour J, Fast P, Bernard R, Ho DD. In vivo electroporation enhances the immunogenicity of an HIV-1 DNA vaccine candidate in healthy volunteers. PLoS One 2011; 6:e19252. [PMID: 21603651 PMCID: PMC3095594 DOI: 10.1371/journal.pone.0019252] [Citation(s) in RCA: 145] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Accepted: 03/23/2011] [Indexed: 11/29/2022] Open
Abstract
Background DNA-based vaccines have been safe but weakly immunogenic in humans to
date. Methods and Findings We sought to determine the safety, tolerability, and immunogenicity of ADVAX,
a multigenic HIV-1 DNA vaccine candidate, injected intramuscularly by
in vivo electroporation (EP) in a Phase-1,
double-blind, randomized placebo-controlled trial in healthy volunteers.
Eight volunteers each received 0.2 mg, 1 mg, or 4 mg ADVAX or saline placebo
via EP, or 4 mg ADVAX via standard intramuscular injection at weeks 0 and 8.
A third vaccination was administered to eleven volunteers at week 36. EP was
safe, well-tolerated and considered acceptable for a prophylactic vaccine.
EP delivery of ADVAX increased the magnitude of HIV-1-specific cell mediated
immunity by up to 70-fold over IM injection, as measured by gamma interferon
ELISpot. The number of antigens to which the response was detected improved
with EP and increasing dosage. Intracellular cytokine staining analysis of
ELISpot responders revealed both CD4+ and CD8+ T cell responses,
with co-secretion of multiple cytokines. Conclusions This is the first demonstration in healthy volunteers that EP is safe,
tolerable, and effective in improving the magnitude, breadth and durability
of cellular immune responses to a DNA vaccine candidate. Trial Registration ClinicalTrials.gov NCT00545987
Collapse
Affiliation(s)
- Sandhya Vasan
- Aaron Diamond AIDS Research Center, New York, New York, United States of America.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
210
|
Safety and efficacy of the HVTN 503/Phambili study of a clade-B-based HIV-1 vaccine in South Africa: a double-blind, randomised, placebo-controlled test-of-concept phase 2b study. THE LANCET. INFECTIOUS DISEASES 2011; 11:507-15. [PMID: 21570355 DOI: 10.1016/s1473-3099(11)70098-6] [Citation(s) in RCA: 288] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND The MRKAd5 HIV-1 gag/pol/nef subtype B vaccine was designed to elicit T-cell-mediated immune responses capable of providing complete or partial protection from HIV-1 infection or a decrease in viral load after acquisition. We aim to assess the safety and efficacy of the vaccine in South Africa, where the major circulating clade is subtype C. METHODS We did a phase 2b double-blind, randomised test-of-concept study in sexually active HIV-1 seronegative participants at five sites in South Africa. Randomisation was by a computer-generated random number sequence. The vaccine and placebo were given by intramuscular injection on a 0, 1, 6 month schedule. Our coprimary endpoints were a vaccine-induced reduction in HIV-1 acquisition and viral-load setpoint. These endpoints were assessed independently in the modified intention-to-treat (MITT) cohort with two-tailed significance tests stratified by sex. We assessed immunogenicity by interferon-γ ELISPOT in peripheral-blood mononuclear cells. After the lack of efficacy of the MRKAd5 HIV-1 vaccine in the Step study, enrolment and vaccination in our study was halted, treatment allocations were unmasked, and follow-up continued. This study is registered with the South Africa National Health Research Database, number DOH-27-0207-1539, and ClinicalTrials.gov, number NCT00413725. FINDINGS 801 of a scheduled 3000 participants, of whom 360 (45%) were women, were randomly assigned to receive either vaccine or placebo. 445 participants (56%) had adenovirus serotype 5 (Ad5) titres greater than 200, and 129 men (29%) were circumcised. 34 MITT participants in the vaccine group were diagnosed with HIV-1 (incidence rate 4·54 per 100 person-years) and 28 in the placebo group (3·70 per 100 person-years). There was no evidence of vaccine efficacy; the hazard ratio adjusted for sex was 1·25 (95% CI 0·76-2·05). Vaccine efficacy did not differ by Ad5 titre, sex, age, herpes simplex virus type 2 status, or circumcision. The geometric mean viral-load setpoint was 20,483 copies per mL (n=33) in the vaccine group and 34,032 copies per mL (n=28) in the placebo group (p=0·39). The vaccine elicited interferon-γ-secreting T cells that recognised both clade B (89%) and C (77%) antigens. INTERPRETATION The MRKAd5 HIV-1 vaccine did not prevent HIV-1 infection or lower viral-load setpoint; however, stopping our trial early probably compromised our ability to draw conclusions. The high incidence rates noted in South Africa highlight the crucial need for intensified efforts to develop an efficacious vaccine. FUNDING The US National Institute of Allergy and Infectious Disease and Merck and Co Inc.
Collapse
|
211
|
Pine SO, Kublin JG, Hammer SM, Borgerding J, Huang Y, Casimiro DR, McElrath MJ. Pre-existing adenovirus immunity modifies a complex mixed Th1 and Th2 cytokine response to an Ad5/HIV-1 vaccine candidate in humans. PLoS One 2011; 6:e18526. [PMID: 21533229 PMCID: PMC3076372 DOI: 10.1371/journal.pone.0018526] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2010] [Accepted: 03/03/2011] [Indexed: 11/30/2022] Open
Abstract
The results of the recent Step Study highlight a need to clarify the effects of pre-existing natural immunity to a vaccine vector on vaccine-induced T-cell responses. To investigate this interaction, we examined the relationship between pre-existing Ad5 immunity and T-cell cytokine response profiles in healthy, HIV-uninfected recipients of MRKAd5 HIV-1 gag vaccine (HVTN 050, ClinicalTrials.gov #NCT00849732). Participants were grouped by baseline Ad5 neutralizing antibody titer as either Ad5-seronegative (titer ≤18; n = 36) or Ad5-seropositive (titer >200; n = 34). Samples from vaccine recipients were analyzed for immune responses to either HIV-1 Gag peptide pools or Ad5 empty vector using an ex vivo assay that measures thirty cytokines in the absence of long-term culture. The overall profiles of cytokine responses to Gag and Ad5 had similar combinations of induced Th1- and Th2-type cytokines, including IFN-γ, IL-2, TNF-α, IP-10, IL-13, and IL-10, although the Ad5-specific responses were uniformly higher than the Gag-specific responses (p<0.0001 for 9 out of 11 significantly expressed analytes). At the peak response time point, PBMC from Ad5-seronegative vaccinees secreted significantly more IP-10 in response to Gag (p = 0.008), and significantly more IP-10 (p = 0.0009), IL-2 (p = 0.006) and IL-10 (p = 0.05) in response to Ad5 empty vector than PBMC from Ad5-seropositive vaccinees. Additionally, similar responses to the Ad5 vector prior to vaccination were observed in almost all subjects, regardless of Ad5 neutralizing antibody status, and the levels of secreted IFN-γ, IL-10, IL-1Ra and GM-CSF were blunted following vaccination. The cytokine response profile of Gag-specific T cells mirrored the Ad5-specific response present in all subjects before vaccination, and included a number of Th1- and Th2-associated cytokines not routinely assessed in current vaccine trials, such as IP-10, IL-10, IL-13, and GM-CSF. Together, these results suggest that vector-specific humoral responses may reduce vaccine-induced T-cell responses by previously undetected mechanisms.
Collapse
Affiliation(s)
- Samuel O. Pine
- Program in Pathobiology, Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - James G. Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Scott M. Hammer
- Division of Infectious Diseases, Columbia University College of Physicians and Surgeons, New York, New York, United States of America
| | - Joleen Borgerding
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Yunda Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Danilo R. Casimiro
- Vaccine Basic Research, Merck Research Laboratories, West Point, Pennsylvania, United States of America
| | - M. Juliana McElrath
- Program in Pathobiology, Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
212
|
Sun C, Zhang Y, Feng L, Pan W, Zhang M, Hong Z, Ma X, Chen X, Chen L. Epidemiology of adenovirus type 5 neutralizing antibodies in healthy people and AIDS patients in Guangzhou, southern China. Vaccine 2011; 29:3837-41. [PMID: 21447314 DOI: 10.1016/j.vaccine.2011.03.042] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Revised: 03/10/2011] [Accepted: 03/12/2011] [Indexed: 10/18/2022]
Abstract
Recombinant adenovirus serotype 5 (Ad5) viruses have been extensively explored as vectors for vaccination or gene therapy. However, one major obstacle to their clinical application is the high prevalence of preexisting anti-Ad5 immunity resulting from natural infection. It has been reported that there are geographic variations in the prevalence of natural adenovirus infection. In the present study, we investigated the seroprevalence of Ad5 in Guangzhou, southern China by measuring the Ad5 neutralizing antibodies in blood samples collected from several sites. The seroprevalence was 77.34% in the general healthy population. The seroprevalence and antibody titers increased with age, with the older population (41-72 years old) having the highest seropositivity (84.8%) and percentage (54.4%) of high Ad5 neutralizing antibody titers (>1000). The dynamics of Ad5 neutralizing antibodies were stable and persistent over the course of eight months. Furthermore, the seroprevalence of Ad5 in the HIV-infected AIDS patients was investigated and there was no significant difference from the general healthy population. Our survey provides useful insights for the future development of Ad5-based vaccination and gene therapy.
Collapse
Affiliation(s)
- Caijun Sun
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Biomedicine and Health (GIBH), Chinese Academy of Sciences, Guangzhou 510530, China
| | | | | | | | | | | | | | | | | |
Collapse
|
213
|
Recombinant adenovirus serotype 26 (Ad26) and Ad35 vaccine vectors bypass immunity to Ad5 and protect nonhuman primates against ebolavirus challenge. J Virol 2011; 85:4222-33. [PMID: 21325402 DOI: 10.1128/jvi.02407-10] [Citation(s) in RCA: 149] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The use of adenoviruses (Ad) as vaccine vectors against a variety of pathogens has demonstrated their capacity to elicit strong antibody and cell-mediated immune responses. Adenovirus serotype C vectors, such as Ad serotype 5 (Ad5), expressing Ebolavirus (EBOV) glycoprotein (GP), protect completely after a single inoculation at a dose of 10(10) viral particles. However, the clinical application of a vaccine based on Ad5 vectors may be hampered, since impairment of Ad5 vaccine efficacy has been demonstrated for humans and nonhuman primates with high levels of preexisting immunity to the vector. Ad26 and Ad35 segregate genetically from Ad5 and exhibit lower seroprevalence in humans, making them attractive vaccine vector alternatives. In the series of studies presented, we show that Ad26 and Ad35 vectors generate robust antigen-specific cell-mediated and humoral immune responses against EBOV GP and that Ad5 immune status does not affect the generation of GP-specific immune responses by these vaccines. We demonstrate partial protection against EBOV by a single-shot Ad26 vaccine and complete protection when this vaccine is boosted by Ad35 1 month later. Increases in efficacy are paralleled by substantial increases in T- and B-cell responses to EBOV GP. These results suggest that Ad26 and Ad35 vectors warrant further development as candidate vaccines for EBOV.
Collapse
|
214
|
Pédron B, Guérin V, Cordeiro DJ, Masmoudi S, Dalle JH, Sterkers G. Development of cytomegalovirus and adenovirus-specific memory CD4 T-cell functions from birth to adulthood. Pediatr Res 2011; 69:106-11. [PMID: 21057376 DOI: 10.1203/pdr.0b013e318204e469] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Age-related changes in memory CD4 T cells (CD4) are poorly known. To address this issue, CD4 proliferative and cytokine responses to an anti-CD3 monoclonal (CD3), to cytomegalovirus (CMV), and to adenovirus (AdV) were assessed in 57 children (age, 0.07-17.16 y) and 17 adults. Results showed i) accumulation of memory CD4 with aging, with 2-3 times more central-memory T cell (TCM; CD45RA/CD62L) than effector-memory T cell (TEM; CD45RA/62L) CD4 at any age. ii) In children older than 2 y, CMV-specific CD4-secreting IFNγ alone predominated over CD4-secreting IL2 + IFNγ and a continuous increase, with aging, in IFNγ responses to the virus was observed. In contrast, in AdV infection, CD4-secreting IL2 + IFNγ predominated and IFNγ responses to the virus reached adult levels from 3 y of age. iii) In children aged 0-2 y, lower total IFNγ responses to CMV (p < 0.02), AdV (p = 0.05), and CD3 (p < 0.01) and lower IFNγ + IL2-responses (p = 0.1, p < 0.02, p < 0.05, respectively) contrasted with no decrease in CD4-secreting IFNγ alone. Defective proliferative responses to AdV (p = 0.03) were also observed. In conclusion, the development of memory CD4 differed in acute AdV and persistent CMV infections. Young age seemed to depress mostly polyfunctional (IL2 + IFNγ secreting) CD4 in both infections.
Collapse
Affiliation(s)
- Béatrice Pédron
- Laboratory of Immunology, Robert Debré Hospital, Paris 75019, France
| | | | | | | | | | | |
Collapse
|
215
|
Krishnapuram R, Kirk-Ballard H, Zuberi A, Dhurandhar NV. Infectivity period of mice inoculated with human adenoviruses. Lab Anim 2011; 45:103-8. [PMID: 21257643 DOI: 10.1258/la.2010.010084] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Due to non-productive infections, mice are not a good model to study some human adenoviruses. However, mice provide an excellent model to study the metabolic effects of human adenovirus, Ad36. Research interest in Ad36 is increasing rapidly, and consequently an increase in the use of mice as a model is anticipated. However, little is known about the transmission potential of Ad36 from infected mice to other laboratory animals or personnel. While underestimating the infectivity could promote inadvertent spread of Ad36, overstating it could drain valuable laboratory resources and animals. Therefore, we determined the duration of infectivity in female C57BL/6J mice that were experimentally infected with human adenoviruses Ad36 or Ad2. Other uninfected mice were co-housed for one week with the experimentally-infected animals, four or eight weeks postinfection. Additionally, uninfected mice were housed in the cages of mice that were infected with Ad36, 12 weeks earlier. The presence of viral DNA in tissues was used to indicate infection of mice. Although experimentally-infected mice harboured viral DNA at least up to 12 weeks, the horizontal transmission of infection was observed in co-housed mice only up to four weeks postinfection. Thus, Ad36-infected mice should be considered potentially infective for eight weeks and appropriate handling and barrier containment should be used. After eight week postinfection, horizontal transmission appears unlikely. This information may provide guidelines for animal handling, and experimental design using Ad36, which may increase safety for laboratory personnel and reduce the number of mice required for experiments.
Collapse
|
216
|
Peiperl L, Morgan C, Moodie Z, Li H, Russell N, Graham BS, Tomaras GD, De Rosa SC, McElrath MJ. Safety and immunogenicity of a replication-defective adenovirus type 5 HIV vaccine in Ad5-seronegative persons: a randomized clinical trial (HVTN 054). PLoS One 2010; 5:e13579. [PMID: 21048953 PMCID: PMC2965084 DOI: 10.1371/journal.pone.0013579] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Accepted: 09/27/2010] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Individuals without prior immunity to a vaccine vector may be more sensitive to reactions following injection, but may also show optimal immune responses to vaccine antigens. To assess safety and maximal tolerated dose of an adenoviral vaccine vector in volunteers without prior immunity, we evaluated a recombinant replication-defective adenovirus type 5 (rAd5) vaccine expressing HIV-1 Gag, Pol, and multiclade Env proteins, VRC-HIVADV014-00-VP, in a randomized, double-blind, dose-escalation, multicenter trial (HVTN study 054) in HIV-1-seronegative participants without detectable neutralizing antibodies (nAb) to the vector. As secondary outcomes, we also assessed T-cell and antibody responses. METHODOLOGY/PRINCIPAL FINDINGS Volunteers received one dose of vaccine at either 10(10) or 10(11) adenovector particle units, or placebo. T-cell responses were measured against pools of global potential T-cell epitope peptides. HIV-1 binding and neutralizing antibodies were assessed. Systemic reactogenicity was greater at the higher dose, but the vaccine was well tolerated at both doses. Although no HIV infections occurred, commercial diagnostic assays were positive in 87% of vaccinees one year after vaccination. More than 85% of vaccinees developed HIV-1-specific T-cell responses detected by IFN-γ ELISpot and ICS assays at day 28. T-cell responses were: CD8-biased; evenly distributed across the three HIV-1 antigens; not substantially increased at the higher dose; and detected at similar frequencies one year following injection. The vaccine induced binding antibodies against at least one HIV-1 Env antigen in all recipients. CONCLUSIONS/SIGNIFICANCE This vaccine appeared safe and was highly immunogenic following a single dose in human volunteers without prior nAb against the vector. TRIAL REGISTRATION ClinicalTrials.gov NCT00119873.
Collapse
Affiliation(s)
- Laurence Peiperl
- University of California San Francisco, San Francisco, California, United States of America
| | - Cecilia Morgan
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Zoe Moodie
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Hongli Li
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Nina Russell
- Bill & Melinda Gates Foundation, Seattle, Washington, United States of America
| | - Barney S. Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | | | - Stephen C. De Rosa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- University of Washington, Seattle, Washington, United States of America
| | - M. Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- University of Washington, Seattle, Washington, United States of America
| | | |
Collapse
|
217
|
Chen H, Xiang ZQ, Li Y, Kurupati RK, Jia B, Bian A, Zhou DM, Hutnick N, Yuan S, Gray C, Serwanga J, Auma B, Kaleebu P, Zhou X, Betts MR, Ertl HCJ. Adenovirus-based vaccines: comparison of vectors from three species of adenoviridae. J Virol 2010; 84:10522-32. [PMID: 20686035 PMCID: PMC2950567 DOI: 10.1128/jvi.00450-10] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Accepted: 07/27/2010] [Indexed: 11/20/2022] Open
Abstract
In order to better understand the broad applicability of adenovirus (Ad) as a vector for human vaccine studies, we compared four adenovirus (Ad) vectors from families C (Ad human serotype 5 [HAdV-5; here referred to as AdHu5]), D (HAdV-26; here referred to as AdHu26), and E (simian serotypes SAdV-23 and SAdV-24; here referred to as chimpanzee serotypes 6 and 7 [AdC6 and AdC7, respectively]) of the Adenoviridae. Seroprevalence rates and titers of neutralizing antibodies to the two human-origin Ads were found to be higher than those reported previously, especially in countries of sub-Saharan Africa. Conversely, prevalence rates and titers to AdC6 and AdC7 were markedly lower. Healthy human adults from the United States had readily detectable circulating T cells recognizing Ad viruses, the levels of which in some individuals were unexpectedly high in response to AdHu26. The magnitude of T-cell responses to AdHu5 correlated with those to AdHu26, suggesting T-cell recognition of conserved epitopes. In mice, all of the different Ad vectors induced CD8(+) T-cell responses that were comparable in their magnitudes and cytokine production profiles. Prime-boost regimens comparing different combinations of Ad vectors failed to indicate that the sequential use of Ad vectors from distinct families resulted in higher immune responses than the use of serologically distinct Ad vectors from the same family. Moreover, the transgene product-specific antibody responses induced by the AdHu26 and AdC vectors were markedly lower than those induced by the AdHu5 vector. AdHu26 vectors and, to a lesser extent, AdC vectors induced more potent Ad-neutralizing antibody responses. These results suggest that the potential of AdHu26 as a vaccine vector may suffer from limitations similar to those found for vectors based on other prevalent human Ads.
Collapse
MESH Headings
- Adenoviridae/classification
- Adenoviridae/genetics
- Adenoviridae/immunology
- Adenoviruses, Human/classification
- Adenoviruses, Human/genetics
- Adenoviruses, Human/immunology
- Adenoviruses, Simian/classification
- Adenoviruses, Simian/genetics
- Adenoviruses, Simian/immunology
- Adult
- Africa South of the Sahara
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Viral/blood
- CD8-Positive T-Lymphocytes/immunology
- CHO Cells
- Capsid/immunology
- Cell Line
- Cricetinae
- Cricetulus
- Female
- Genetic Vectors
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Humans
- Mice
- Mice, Inbred BALB C
- Rabies virus/immunology
- Receptors, Virus/metabolism
- Recombinant Proteins/genetics
- Recombinant Proteins/metabolism
- Seroepidemiologic Studies
- Serotyping
- Species Specificity
- Viral Vaccines/genetics
Collapse
Affiliation(s)
- H. Chen
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Z. Q. Xiang
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Y. Li
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - R. K. Kurupati
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - B. Jia
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - A. Bian
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - D. M. Zhou
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - N. Hutnick
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - S. Yuan
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - C. Gray
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - J. Serwanga
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - B. Auma
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - P. Kaleebu
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - X. Zhou
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - M. R. Betts
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - H. C. J. Ertl
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
218
|
Ranki T, Hemminki A. Serotype chimeric human adenoviruses for cancer gene therapy. Viruses 2010; 2:2196-2212. [PMID: 21994616 PMCID: PMC3185575 DOI: 10.3390/v2102196] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Revised: 08/16/2010] [Accepted: 09/22/2010] [Indexed: 11/16/2022] Open
Abstract
Cancer gene therapy consists of numerous approaches where the common denominator is utilization of vectors for achieving therapeutic effect. A particularly potent embodiment of the approach is virotherapy, in which the replication potential of an oncolytic virus is directed towards tumor cells to cause lysis, while normal cells are spared. Importantly, the therapeutic effect of the initial viral load is amplified through viral replication cycles and production of progeny virions. All cancer gene therapy approaches rely on a sufficient level of delivery of the anticancer agent into target cells. Thus, enhancement of delivery to target cells, and reduction of delivery to non-target cells, in an approach called transductional targeting, is attractive. Both genetic and non-genetic retargeting strategies have been utilized. However, in the context of oncolytic viruses, it is beneficial to have the specific modification included in progeny virions and hence genetic modification may be preferable. Serotype chimerism utilizes serotype specific differences in receptor usage, liver tropism and seroprevalence in order to gain enhanced infection of target tissue. This review will focus on serotype chimeric adenoviruses for cancer gene therapy applications.
Collapse
Affiliation(s)
- Tuuli Ranki
- Cancer Gene Therapy Group, Molecular Cancer Biology Program, University of Helsinki, P.O. Box 63, 00014 University of Helsinki, Finland; E-Mail:
- HUSLAB, Helsinki University Central Hospital, P.O. Box 100, 00029 HUS, Helsinki, Finland
- Haartman Institute & Transplantation Laboratory, University of Helsinki, P.O. Box 21, 00014 University of Helsinki, Finland
- Finnish Institute for Molecular Medicine, University of Helsinki, P.O.Box 20, 00014 University of Helsinki, Finland
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Molecular Cancer Biology Program, University of Helsinki, P.O. Box 63, 00014 University of Helsinki, Finland; E-Mail:
- HUSLAB, Helsinki University Central Hospital, P.O. Box 100, 00029 HUS, Helsinki, Finland
- Haartman Institute & Transplantation Laboratory, University of Helsinki, P.O. Box 21, 00014 University of Helsinki, Finland
- Finnish Institute for Molecular Medicine, University of Helsinki, P.O.Box 20, 00014 University of Helsinki, Finland
- Author to whom correspondence should be addressed; E-Mail: ; Tel. +358-9-1912 5464; Fax: +358-9-1912 5465
| |
Collapse
|
219
|
Jaoko W, Karita E, Kayitenkore K, Omosa-Manyonyi G, Allen S, Than S, Adams EM, Graham BS, Koup RA, Bailer RT, Smith C, Dally L, Farah B, Anzala O, Muvunyi CM, Bizimana J, Tarragona-Fiol T, Bergin PJ, Hayes P, Ho M, Loughran K, Komaroff W, Stevens G, Thomson H, Boaz MJ, Cox JH, Schmidt C, Gilmour J, Nabel GJ, Fast P, Bwayo J. Safety and immunogenicity study of Multiclade HIV-1 adenoviral vector vaccine alone or as boost following a multiclade HIV-1 DNA vaccine in Africa. PLoS One 2010; 5:e12873. [PMID: 20877623 PMCID: PMC2943475 DOI: 10.1371/journal.pone.0012873] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Accepted: 08/23/2010] [Indexed: 11/21/2022] Open
Abstract
Background We conducted a double-blind, randomized, placebo-controlled Phase I study of a recombinant replication-defective adenovirus type 5 (rAd5) vector expressing HIV-1 Gag and Pol from subtype B and Env from subtypes A, B and C, given alone or as boost following a DNA plasmid vaccine expressing the same HIV-1 proteins plus Nef, in 114 healthy HIV-uninfected African adults. Methodology/Principal Findings Volunteers were randomized to 4 groups receiving the rAd5 vaccine intramuscularly at dosage levels of 1×1010 or 1×1011 particle units (PU) either alone or as boost following 3 injections of the DNA vaccine given at 4 mg/dose intramuscularly by needle-free injection using Biojector® 2000. Safety and immunogenicity were evaluated for 12 months. Both vaccines were well-tolerated. Overall, 62% and 86% of vaccine recipients in the rAd5 alone and DNA prime - rAd5 boost groups, respectively, responded to the HIV-1 proteins by an interferon-gamma (IFN-γ) ELISPOT. The frequency of immune responses was independent of rAd5 dosage levels. The highest frequency of responses after rAd5 alone was detected at 6 weeks; after DNA prime - rAd5 boost, at 6 months (end of study). At baseline, neutralizing antibodies against Ad5 were present in 81% of volunteers; the distribution was similar across the 4 groups. Pre-existing immunity to Ad5 did not appear to have a significant impact on reactogenicity or immune response rates to HIV antigens by IFN-γ ELISPOT. Binding antibodies against Env were detected in up to 100% recipients of DNA prime - rAd5 boost. One volunteer acquired HIV infection after the study ended, two years after receipt of rAd5 alone. Conclusions/Significance The HIV-1 rAd5 vaccine, either alone or as a boost following HIV-1 DNA vaccine, was well-tolerated and immunogenic in African adults. DNA priming increased the frequency and magnitude of cellular and humoral immune responses, but there was no effect of rAd5 dosage on immunogenicity endpoints. Trial Registration ClinicalTrials.gov NCT00124007
Collapse
MESH Headings
- AIDS Vaccines/adverse effects
- AIDS Vaccines/immunology
- Adenoviridae/genetics
- Adenoviridae/immunology
- Adolescent
- Adult
- Antibodies, Viral/immunology
- Double-Blind Method
- Drug-Related Side Effects and Adverse Reactions
- Genetic Vectors/adverse effects
- Genetic Vectors/genetics
- Genetic Vectors/immunology
- HIV Infections/immunology
- HIV Infections/prevention & control
- HIV Infections/virology
- HIV-1/classification
- HIV-1/genetics
- HIV-1/immunology
- Humans
- Immunization, Secondary
- Male
- Middle Aged
- Vaccines, DNA/adverse effects
- Vaccines, DNA/immunology
- Young Adult
- gag Gene Products, Human Immunodeficiency Virus/adverse effects
- gag Gene Products, Human Immunodeficiency Virus/genetics
- gag Gene Products, Human Immunodeficiency Virus/immunology
- pol Gene Products, Human Immunodeficiency Virus/adverse effects
- pol Gene Products, Human Immunodeficiency Virus/genetics
- pol Gene Products, Human Immunodeficiency Virus/immunology
Collapse
Affiliation(s)
- Walter Jaoko
- Kenya AIDS Vaccine Initiative (KAVI), Nairobi, Kenya
| | - Etienne Karita
- Projet San Francisco (PSF), Rwanda-Zambia HIV Research Project, Kigali, Rwanda
| | | | | | - Susan Allen
- Rollins School of Public Health, Emory University, Atlanta, Georgia, United States of America
| | - Soe Than
- International AIDS Vaccine Initiative (IAVI), New York, New York, United States of America
| | - Elizabeth M. Adams
- Vaccine Clinical Research Branch (VCRB), Vaccine Research Program (VRP)/Division of AIDS (DAIDS)/National Institute of Allergy and Infectious Diseases (NIAID)/National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Barney S. Graham
- Vaccine Research Center (VRC)/NIAID/NIH, Bethesda, Maryland, United States of America
| | - Richard A. Koup
- Vaccine Research Center (VRC)/NIAID/NIH, Bethesda, Maryland, United States of America
| | - Robert T. Bailer
- Vaccine Research Center (VRC)/NIAID/NIH, Bethesda, Maryland, United States of America
| | - Carol Smith
- The EMMES Corporation, Rockville, Maryland, United States of America
| | - Len Dally
- The EMMES Corporation, Rockville, Maryland, United States of America
| | - Bashir Farah
- Kenya AIDS Vaccine Initiative (KAVI), Nairobi, Kenya
| | - Omu Anzala
- Kenya AIDS Vaccine Initiative (KAVI), Nairobi, Kenya
| | - Claude M. Muvunyi
- Projet San Francisco (PSF), Rwanda-Zambia HIV Research Project, Kigali, Rwanda
| | - Jean Bizimana
- Projet San Francisco (PSF), Rwanda-Zambia HIV Research Project, Kigali, Rwanda
| | | | - Philip J. Bergin
- IAVI Human Immunology Laboratory, Imperial College, London, United Kingdom
| | - Peter Hayes
- IAVI Human Immunology Laboratory, Imperial College, London, United Kingdom
| | - Martin Ho
- The EMMES Corporation, Rockville, Maryland, United States of America
| | - Kelley Loughran
- The EMMES Corporation, Rockville, Maryland, United States of America
| | - Wendy Komaroff
- International AIDS Vaccine Initiative (IAVI), New York, New York, United States of America
| | - Gwynneth Stevens
- International AIDS Vaccine Initiative (IAVI), New York, New York, United States of America
| | - Helen Thomson
- International AIDS Vaccine Initiative (IAVI), New York, New York, United States of America
| | - Mark J. Boaz
- International AIDS Vaccine Initiative (IAVI), New York, New York, United States of America
| | - Josephine H. Cox
- International AIDS Vaccine Initiative (IAVI), New York, New York, United States of America
| | - Claudia Schmidt
- International AIDS Vaccine Initiative (IAVI), New York, New York, United States of America
| | - Jill Gilmour
- IAVI Human Immunology Laboratory, Imperial College, London, United Kingdom
| | - Gary J. Nabel
- Vaccine Research Center (VRC)/NIAID/NIH, Bethesda, Maryland, United States of America
| | - Patricia Fast
- International AIDS Vaccine Initiative (IAVI), New York, New York, United States of America
- * E-mail:
| | - Job Bwayo
- Kenya AIDS Vaccine Initiative (KAVI), Nairobi, Kenya
| |
Collapse
|
220
|
Neutralizing antibodies to human and simian adenoviruses in humans and New-World monkeys. Virology 2010; 407:1-6. [PMID: 20797754 DOI: 10.1016/j.virol.2010.07.043] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Revised: 05/13/2010] [Accepted: 07/28/2010] [Indexed: 11/24/2022]
Abstract
Vaccines based on adenovirus (Ad) vectors are currently in development against several pathogens. However, neutralizing antibodies (NAb) to human adenovirus type 5 (AdHu5), the best-studied vector, are highly prevalent in humans worldwide. Less-prevalent adenoviruses, including human and simian serotypes, provide alternative vaccine platforms. In this study, sera from 200 Brazilian human subjects and New-World monkeys were tested for NAb titers to human serotypes AdHu5 and AdHu26 and chimpanzee-origin Ad viruses of serotype 6 (AdC6) and serotype 68 (AdC68). Seroprevalence rates of NAb in humans were 69.5% for AdHu5, 44% for AdHu26, 21% for AdC6 and 23.5% for AdC68. In addition, NAb titers to human Ad were consistently higher than those found to simian serotypes. Surprisingly, sera from some New-World monkey species were able to neutralize AdC6 and/or AdC68. A possible explanation for these findings and the implications for the development of Ad-vector vaccines are discussed in detail.
Collapse
|
221
|
Stephen SL, Freestone K, Dunn S, Twigg MW, Homer-Vanniasinkam S, Walker JH, Wheatcroft SB, Ponnambalam S. Scavenger receptors and their potential as therapeutic targets in the treatment of cardiovascular disease. Int J Hypertens 2010; 2010:646929. [PMID: 20981357 PMCID: PMC2958427 DOI: 10.4061/2010/646929] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2010] [Accepted: 07/07/2010] [Indexed: 12/12/2022] Open
Abstract
Scavenger receptors act as membrane-bound and soluble proteins that bind to macromolecular complexes and pathogens. This diverse supergroup of proteins mediates binding to modified lipoprotein particles which regulate the initiation and progression of atherosclerotic plaques. In vascular tissues, scavenger receptors are implicated in regulating intracellular signaling, lipid accumulation, foam cell development, and cellular apoptosis or necrosis linked to the pathophysiology of atherosclerosis. One approach is using gene therapy to modulate scavenger receptor function in atherosclerosis. Ectopic expression of membrane-bound scavenger receptors using viral vectors can modify lipid profiles and reduce the incidence of atherosclerosis. Alternatively, expression of soluble scavenger receptors can also block plaque initiation and progression. Inhibition of scavenger receptor expression using a combined gene therapy and RNA interference strategy also holds promise for long-term therapy. Here we review our current understanding of the gene delivery by viral vectors to cells and tissues in gene therapy strategies and its application to the modulation of scavenger receptor function in atherosclerosis.
Collapse
Affiliation(s)
- Sam L Stephen
- Endothelial Cell Biology Unit, Institute of Molecular & Cellular Biology, LIGHT Laboratories, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
| | | | | | | | | | | | | | | |
Collapse
|
222
|
Mastelic B, Ahmed S, Egan WM, Del Giudice G, Golding H, Gust I, Neels P, Reed SG, Sheets RL, Siegrist CA, Lambert PH. Mode of action of adjuvants: implications for vaccine safety and design. Biologicals 2010; 38:594-601. [PMID: 20659806 DOI: 10.1016/j.biologicals.2010.06.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Accepted: 06/27/2010] [Indexed: 11/18/2022] Open
Abstract
For decades, the search for new vaccine adjuvants has been largely empirical. A series of new adjuvants and related formulations are now emerging that are acting through identified immunological mechanisms. Understanding adjuvant mechanism of action is crucial for vaccine design, since this allows for directing immune responses towards efficacious disease-specific effector mechanisms and appropriate memory. It is also of great importance to build new paradigms for assessing adjuvant safety at development stages and at regulatory level. This report reflects the conclusions of a group of scientists from academia, regulatory agencies and industry who attended a conference, organized by the International Association for Biologicals (IABS), on the mode of action of adjuvants on 29-30 April 2010 in Bethesda, Maryland, USA, particularly focusing on how understanding adjuvants mode of action can impact on the assessment of vaccine safety and help to develop target-specific vaccines. More information on the conference output can be found on the IABS website, http://www.iabs.org/.
Collapse
Affiliation(s)
- Béatris Mastelic
- WHO-Center for Vaccinology and Neonatal Immunology, CMU, 1 rue Michel-Servet, 1211 Geneva, 4, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
223
|
Biodistribution and retargeting of FX-binding ablated adenovirus serotype 5 vectors. Blood 2010; 116:2656-64. [PMID: 20610817 DOI: 10.1182/blood-2009-12-260026] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A major limitation for adenoviral transduction in vivo is the profound liver tropism of adenovirus type 5 (Ad5). Recently, we demonstrated that coagulation factor X (FX) binds to Ad5-hexon protein at high affinity to mediate hepatocyte transduction after intravascular delivery. We developed novel genetically FX-binding ablated Ad5 vectors with lower liver transduction. Here, we demonstrate that FX-binding ablated Ad5 predominantly localize to the liver and spleen 1 hour after injection; however, they had highly reduced liver transduction in both control and macrophage-depleted mice compared with Ad5. At high doses in macrophage-depleted mice, FX-binding ablated vectors transduced the spleen more efficiently than Ad5. Immunohistochemical studies demonstrated transgene colocalization with CD11c(+), ER-TR7(+), and MAdCAM-1(+) cells in the splenic marginal zone. Systemic inflammatory profiles were broadly similar between FX-binding ablated Ad5 and Ad5 at low and intermediate doses, although higher levels of several inflammatory proteins were observed at the highest dose of FX-binding ablated Ad5. Subsequently, we generated a FX-binding ablated virus containing a high affinity Ad35 fiber that mediated a significant improvement in lung/liver ratio in macrophage-depleted CD46(+) mice compared with controls. Therefore, this study documents the biodistribution and reports the retargeting capacity of FX binding-ablated Ad5 vectors in vitro and in vivo.
Collapse
|
224
|
Gabitzsch ES, Xu Y, Balint JP, Hartman ZC, Lyerly HK, Jones FR. Anti-tumor immunotherapy despite immunity to adenovirus using a novel adenoviral vector Ad5 [E1-, E2b-]-CEA. Cancer Immunol Immunother 2010; 59:1131-5. [PMID: 20361185 PMCID: PMC11030277 DOI: 10.1007/s00262-010-0847-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Accepted: 03/10/2010] [Indexed: 10/19/2022]
Abstract
Adenovirus serotype 5 (Ad5) has been widely used in clinical trials because it expresses inserted transgenes robustly and augments the innate immune response. Strategies to improve Ad5 vectors that can circumvent Ad5 immunity have become a critical issue, especially for use as a cancer immunotherapeutic in which repeated immunization is required. In this study, we constructed a novel Ad5 vector with unique deletions of the viral DNA polymerase and the pre-terminal protein region (Ad5 [E1-, E2b-]). This vector contains the carcinoembryonic antigen (CEA) gene insert and is designed to induce cell-mediated immunity (CMI) against the tumor-associated target. The CEA immunogenicity and in vivo anti-tumor effects of repeated immunizations with Ad5 [E1-, E2b-]-CEA compared with those observed with current generation Ad5 [E1-]-CEA were tested in Ad5 pre-immunized mice. We report that Ad5-immune mice immunized multiple times with Ad5 [E1-, E2b-]-CEA induced CEA-specific CMI responses that were significantly increased over those detected in Ad5-immune mice immunized multiple times with a current generation Ad5 [E1-]-CEA. Ad5 immune mice bearing CEA-expressing tumors that were treated with Ad5 [E1-, E2b-]-CEA had increased anti-tumor response as compared with Ad5 [E1-]-CEA treated mice. These results demonstrate that Ad5 [E1-, E2b-]-CEA can induce CMI immune responses which result in tumor growth inhibition despite the presence of pre-existing Ad5 immunity. Multiple re-immunizations using the same vector platform are now possible with the novel Ad5 [E1-, E2b-] platform.
Collapse
|
225
|
Lung homing CTLs and their proliferation ability are important correlates of vaccine protection against influenza. Vaccine 2010; 28:5669-75. [PMID: 20600493 DOI: 10.1016/j.vaccine.2010.06.053] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Revised: 06/09/2010] [Accepted: 06/15/2010] [Indexed: 01/17/2023]
Abstract
We conducted a study to evaluate the protective efficacy in mice of vaccination with novel adenovirus vectors expressing an influenza A nucleoprotein (AdFluA-NP) based on isolates from non-human primates. In a previous study, we had observed that AdFluA-NP vectors can induce similar T cell responses in mice yet differ in ability to protect animals from lethal challenge with influenza A virus. To better define correlates of protection, we extended our study design to include additional novel AdFluA-NP vectors, and to evaluate cytotoxic T lymphocyte (CTL) responses in the spleens and lungs of immunized mice prior to virus challenge. As in our previous study, all vectors induced similar numbers of antigen-specific interferon gamma (IFNgamma) secreting T cells and memory T cells in the spleen 4 weeks post immunization, but differed in their ability to protect the animals from lethal infection. However, cytokine-secreting NP antigen-specific CTLs in the lungs of mice from immunization groups that survived lethal challenge showed greater proliferative ability and higher CD27 expression. In addition, NP antigen-specific peripheral blood lymphocytes from protected mice showed greater proliferative ability after ex vivo stimulation. Our results provide additional correlates of protection that should be considered when developing anti-influenza vaccines.
Collapse
|
226
|
Frequent detection of human adenovirus from the lower gastrointestinal tract in men who have sex with men. PLoS One 2010; 5:e11321. [PMID: 20593015 PMCID: PMC2892480 DOI: 10.1371/journal.pone.0011321] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Accepted: 05/01/2010] [Indexed: 12/19/2022] Open
Abstract
Background The association between baseline seropositivity to human adenovirus (HAdV) type 5 and increased HIV acquisition in the Step HIV Vaccine Study has raised questions concerning frequency of acquired and/or persistent Adenovirus infections among adults at high risk of HIV-1 infection. Methodology To evaluate the frequency and pattern of HAdV shedding from the lower GI tract, we retrospectively tested rectal swabs for HAdVs in a cohort of 20 HSV-2 positive HIV-positive Peruvian men who have sex with men (MSM) undergoing rectal swabbing three times/week for 18 consecutive weeks, in a prospective study of HSV-2 suppression in HIV infection. Viral DNA was extracted and amplified using a sensitive multiplex PCR assay that detects all currently recognized HAdV types. Molecular typing of viruses was performed on selected samples by hexon gene sequencing. Baseline neutralizing antibody titers to HAdVs −5, −26, −35 and −48 were also assessed. Principal Findings 15/20 individuals had HAdV detected during follow up. The median frequency of HAdV detection was 30% of samples (range 2.0% to 64.7%). HAdV shedding typically occurred on consecutive days in clustered episodes lasting a median of 4 days (range 1 to 9 days) separated by periods without shedding, suggesting frequent new infections or reactivation of latent infections over time. 8 of the 15 shedders had more than one type detected in follow-up. 20 HAdV types from species B, C, and D were identified, including HAdV-5, −26 and −48, HAdV types under development as potential vaccine candidates. 14/20 subjects were seropositive for HAdV-5; 15/20 for HAdV-26; 3/20 for HAdV-35; and 2/20 for HAdV-48. HAdV shedding did not correlate with CD4 count, plasma HIV-1 viral load, or titers to HAdV-5 or HAdV-35. The sole individual with HAdV-5 shedding was HAdV-5 seropositive. Conclusions HAdV shedding was highly prevalent and diverse, including types presently under consideration as HIV vaccine vectors. Subclinical HAdV infection of the GI tract is common among MSM in Peru; the prevalence of HAdV in the enteric tract should be evaluated in other populations. The association between ongoing recent enteric HAdV and the immune response to recombinant HAdV vaccines should be evaluated.
Collapse
|