1
|
Zhang Y, Klein K, Ratcliff A, Galappaththi SL, Hathaway N, Twells N, Patel M, Temesy S, Bailey J, Mahal L, Creuzenet C, Arts E. Transmitted/founder (T/F) HIV-1 derived from sexual contact exhibits greater transmission fitness in human cervical tissue than T/F HIV-1 from blood-to-blood contact: Unique glycan profiles on T/F envelopes associated with transmission phenotypes. PLoS Pathog 2025; 21:e1013177. [PMID: 40408432 DOI: 10.1371/journal.ppat.1013177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 05/01/2025] [Indexed: 05/25/2025] Open
Abstract
Human immunodeficiency virus 1 (HIV-1) risk groups include, but are not limited to, heterosexual individuals (HET), men-who-have-sex-with-men (MSM), and people who inject drugs (PWID). Although genetically diverse HIV-1 populations are transferred from donor to recipient, systemic infection is often established by a single clone, the transmitted/founder (T/F) virus. This phenomenon is especially prevalent in sexual transmission, but less stringent in blood-to-blood contact transmission. Specific traits that permit successful transmission have not been well characterized. Thus, HIV-1 containing the chimeric T/F envelope (Env) from different transmission routes was assessed for ex vivo transmission fitness by performing mixed competition assays (also referred to as mixed competitions) on human cervical tissues. We found that chimeric T/F viruses isolated from the PWID exhibit limited replication capacity in cervical tissues when compared to those from MSM and HET, diminishing their chances of transmission to T helper type 1 (Th1) and Th17 cells. This reduced transmission fitness of T/F HIV-1 from PWID was not observed when infecting Th1 and Th17 cells directly, bypassing cervical tissues. Phenotypic assays showed that the chimeric T/F viruses from PWID differed from other groups by having an enhanced ability to utilize diverse CCR5 conformations, while Env expression level, CD4/CCR5 utilization, and entry speed did not differ. Different glycosylation profiles were detected on T/F compared to chronic Env with increased complex, fucosylated N- and O-glycans found more frequently on the T/F Env. Furthermore, the increased presence of these fucosylated glycans correlated with replication fitness in cervical tissues. In contrast, bisecting branched N-glycan found more frequently on chronic Env was associated with decreased entry efficiency and more stringent usage of CCR5. These findings suggest that glycosylation patterns/levels and/or Env structure greatly impact the differences in transmission fitness of T/F HIV-1.
Collapse
Affiliation(s)
- Yiying Zhang
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
| | - Katja Klein
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
- Bristol Veterinary School, University of Bristol, Bristol, United Kingdom
| | - Annette Ratcliff
- Department of Molecular Biology and Microbiology and Division of Infectious Diseases, Case Western Reserve University, Cleveland, United States of America
| | | | - Nicholas Hathaway
- Department of Pathology and Laboratory Medicine, Brown University, Providence, United States of America
| | - Nicholas Twells
- Department of Chemistry, University of Alberta, Edmonton, Canada
| | - Mukti Patel
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
| | - Stephen Temesy
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
| | - Jeffrey Bailey
- Department of Pathology and Laboratory Medicine, Brown University, Providence, United States of America
| | - Lara Mahal
- Department of Chemistry, University of Alberta, Edmonton, Canada
| | - Carole Creuzenet
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
| | - Eric Arts
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
- Department of Molecular Biology and Microbiology and Division of Infectious Diseases, Case Western Reserve University, Cleveland, United States of America
| |
Collapse
|
2
|
Tarlinton DM, Ding Z, Tellier J, Nutt SL. Making sense of plasma cell heterogeneity. Curr Opin Immunol 2023; 81:102297. [PMID: 36889029 DOI: 10.1016/j.coi.2023.102297] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/26/2023] [Accepted: 01/29/2023] [Indexed: 03/08/2023]
Abstract
Plasma cells (PCs) are essential for the quality and longevity of protective immunity. The canonical humoral response to vaccination involves induction of germinal centers in lymph nodes followed by maintenance by bone marrow-resident PCs, although there are many variations of this theme. Recent studies have highlighted the importance of PCs in nonlymphoid organs, including the gut, central nervous system, and skin. These sites harbor PCs with distinct isotypes and possible immunoglobulin-independent functions. Indeed, bone marrow now appears unique in housing PCs derived from multiple other organs. The mechanisms through which the bone marrow maintains PC survival long-term and the impact of their diverse origins on this process remain very active areas of research.
Collapse
Affiliation(s)
- David M Tarlinton
- Department of Immunology, Monash University, Melbourne, Victoria, Australia.
| | - Zhoujie Ding
- Department of Immunology, Monash University, Melbourne, Victoria, Australia
| | - Julie Tellier
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Stephen L Nutt
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
3
|
Abstract
Epithelial barriers, which include the gastrointestinal, respiratory, and genitourinary mucosa, compose the body’s front line of defense. Since barrier tissues are persistently exposed to microbial challenges, a rapid response that can deal with diverse invading pathogens is crucial. Because B cells have been perceived as indirectly contributing to immune responses through antibody production, B cells functioning in the peripheral organs have been outside the scope of researchers. However, recent evidence supports the existence of tissue-resident memory B cells (BRMs) in the lungs. This population’s defensive response was stronger and faster than that of their circulating counterparts and could resist heterogeneous strains. With such traits, BRMs could be a promising target for vaccine design, but much about them remains to be revealed, including their locations, origin, specific markers, and the mechanisms of their establishment and maintenance. There is evidence for resident B cells in organs other than the lungs, suggesting that B cells are directly involved in the immune reactions of multiple non-lymphoid organs. This review summarizes the history of the discovery of BRMs and discusses important unresolved questions. Unique characteristics of humoral immunity that play an important role in the peripheral organs will be described briefly. Future research on B cells residing in non-lymphoid organs will provide new insights to help solve major problems regarding human health.
Collapse
Affiliation(s)
- Choong Man Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Ji Eun Oh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- BioMedical Research Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- *Correspondence: Ji Eun Oh,
| |
Collapse
|
4
|
Sobia P, Archary D. Preventive HIV Vaccines-Leveraging on Lessons from the Past to Pave the Way Forward. Vaccines (Basel) 2021; 9:vaccines9091001. [PMID: 34579238 PMCID: PMC8472969 DOI: 10.3390/vaccines9091001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 12/05/2022] Open
Abstract
Almost four decades on, since the 1980’s, with hundreds of HIV vaccine candidates tested in both non-human primates and humans, and several HIV vaccines trials later, an efficacious HIV vaccine continues to evade us. The enormous worldwide genetic diversity of HIV, combined with HIV’s inherent recombination and high mutation rates, has hampered the development of an effective vaccine. Despite the advent of antiretrovirals as pre-exposure prophylaxis and preventative treatment, which have shown to be effective, HIV infections continue to proliferate, highlighting the great need for a vaccine. Here, we provide a brief history for the HIV vaccine field, with the most recent disappointments and advancements. We also provide an update on current passive immunity trials, testing proof of the concept of the most clinically advanced broadly neutralizing monoclonal antibodies for HIV prevention. Finally, we include mucosal immunity, the importance of vaccine-elicited immune responses and the challenges thereof in the most vulnerable environment–the female genital tract and the rectal surfaces of the gastrointestinal tract for heterosexual and men who have sex with men transmissions, respectively.
Collapse
Affiliation(s)
- Parveen Sobia
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa;
| | - Derseree Archary
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa;
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban 4001, South Africa
- Correspondence: ; Tel.: +27-(0)-31-655-0540
| |
Collapse
|
5
|
Gehlhausen JR, Iwasaki A. B cells join T cell clusters in the host response to recurrent herpes simplex virus 2 infection. J Clin Invest 2021; 131:148300. [PMID: 33938452 DOI: 10.1172/jci148300] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Recurrent genital herpes lesions are infiltrated by various leukocytes, yet the role of B cell subsets in this process is unknown. In this issue of the JCI, Ford et al. describe the presence and antibody-secreting role of local B cell populations in herpes simplex virus 2 (HSV-2) recurrent lesions. The authors analyzed a comprehensive array of sequential skin biopsy specimens from HSV-2-infected patients over time and at various stages of infection. Using immunofluorescence and in situ hybridization, the authors show the presence of rare IgD+ naive B cells and IgG-expressing antibody-secreting cells (ASCs) in recurrent HSV-2 lesions embedded in CD4+ T cell-rich dermal immune infiltrates, levels of which transiently increase during lesion reactivation and healing. Notably, local increases in HSV-2-specific antibodies in recurrent lesions were detected, whereas serum HSV-2 antibody levels remained stable. Future research is needed to understand the precise role of these tissue-visiting B cells in disease resolution.
Collapse
Affiliation(s)
| | - Akiko Iwasaki
- Department of Dermatology and.,Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Molecular Cellular and Developmental Biology, Yale University, New Haven, Connecticut, USA.,Howard Hughes Medical Institute
| |
Collapse
|
6
|
Logerot S, Figueiredo-Morgado S, Charmeteau-de-Muylder B, Sandouk A, Drillet-Dangeard AS, Bomsel M, Bourgault-Villada I, Couëdel-Courteille A, Cheynier R, Rancez M. IL-7-Adjuvanted Vaginal Vaccine Elicits Strong Mucosal Immune Responses in Non-Human Primates. Front Immunol 2021; 12:614115. [PMID: 33717097 PMCID: PMC7947860 DOI: 10.3389/fimmu.2021.614115] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 01/11/2021] [Indexed: 12/26/2022] Open
Abstract
Mucosal immune responses are crucial in protecting against pathogens entering through mucosal surfaces. However, due to poor T-cell responsiveness upon mucosal antigenic stimulation, mucosal immunity remains difficult to obtain through vaccines and requires appropriate adjuvants. We previously demonstrated that administered systemically to healthy macaques or locally expressed in the intestinal mucosa of acutely SIV-infected macaques, interleukin-7 (IL-7) triggers chemokine expression and immune cell homing into mucosae, suggesting its important role in the development of mucosal immune responses. We therefore examined whether local delivery of recombinant glycosylated simian IL-7 (rs-IL-7gly) to the vaginal mucosa of rhesus macaques could prepare the lower female genital tract (FGT) for subsequent immunization and act as an efficient mucosal adjuvant. First, we showed that local administration of rs-IL-7gly triggers vaginal overexpression of chemokines and infiltration of mDCs, macrophages, NKs, B- and T-cells in the lamina propria while MamuLa-DR+ APCs accumulated in the epithelium. Subsequent mucosal anti-DT immunization in macaques resulted in a faster, stronger, and more persistent mucosal antibody response compared to DT-immunization alone. Indeed, we detected robust productions of DT-specific IgAs and IgGs in their vaginal secretions and identified cells secreting DT-specific IgAs in their vaginal mucosa and IgGs in draining lymph nodes. Finally, the expression of chemokines involved in the organization of tertiary lymphoid structures (TLS) was only increased in the vaginal mucosa of IL-7-adjuvanted immunized macaques. Interestingly, TLSs developed around PNAd+ high endothelial venules in their lower FGT sampled 2 weeks after the last immunization. Non-traumatic vaginal administration of rs-IL-7gly prepares the mucosa to respond to subsequent local immunization and allows the development of a strong mucosal immune response in macaques, through the chemokine-dependent recruitment of immune cells, the activation of mDCs and the formation of TLSs. The localization of DT-specific IgA+ plasma cells in the upper vaginal mucosa argues for their contribution to the production of specific immunoglobulins in the vaginal secretions. Our results highlight the potential of IL-7 as a potent mucosal adjuvant to stimulate the FGT immune system and elicit vaginal antibody responses to local immunization, which is the most promising way to confer protection against many sexually transmitted diseases.
Collapse
Affiliation(s)
- Sandrine Logerot
- Laboratory of Dendritic Cells, B Lymphocytes and Cytokines in their Microenvironment During Viral Infections and Cancer, Department of Infection, Immunity and Inflammation, Université de Paris, INSERM, CNRS, Institut Cochin, Paris, France
| | - Suzanne Figueiredo-Morgado
- Laboratory of Dendritic Cells, B Lymphocytes and Cytokines in their Microenvironment During Viral Infections and Cancer, Department of Infection, Immunity and Inflammation, Université de Paris, INSERM, CNRS, Institut Cochin, Paris, France
| | - Bénédicte Charmeteau-de-Muylder
- Laboratory of Dendritic Cells, B Lymphocytes and Cytokines in their Microenvironment During Viral Infections and Cancer, Department of Infection, Immunity and Inflammation, Université de Paris, INSERM, CNRS, Institut Cochin, Paris, France
| | - Abdelkader Sandouk
- Laboratory of Dendritic Cells, B Lymphocytes and Cytokines in their Microenvironment During Viral Infections and Cancer, Department of Infection, Immunity and Inflammation, Université de Paris, INSERM, CNRS, Institut Cochin, Paris, France
| | - Anne-Sophie Drillet-Dangeard
- Laboratory of Mucosal Entry of HIV-1 and Mucosal Immunity, Department of Infection, Immunity and Inflammation, Université de Paris, INSERM, CNRS, Institut Cochin, Paris, France
| | - Morgane Bomsel
- Laboratory of Mucosal Entry of HIV-1 and Mucosal Immunity, Department of Infection, Immunity and Inflammation, Université de Paris, INSERM, CNRS, Institut Cochin, Paris, France
| | - Isabelle Bourgault-Villada
- Laboratory of Dendritic Cells, B Lymphocytes and Cytokines in their Microenvironment During Viral Infections and Cancer, Department of Infection, Immunity and Inflammation, Université de Paris, INSERM, CNRS, Institut Cochin, Paris, France
| | - Anne Couëdel-Courteille
- Laboratory of Dendritic Cells, B Lymphocytes and Cytokines in their Microenvironment During Viral Infections and Cancer, Department of Infection, Immunity and Inflammation, Université de Paris, INSERM, CNRS, Institut Cochin, Paris, France
| | - Rémi Cheynier
- Laboratory of Dendritic Cells, B Lymphocytes and Cytokines in their Microenvironment During Viral Infections and Cancer, Department of Infection, Immunity and Inflammation, Université de Paris, INSERM, CNRS, Institut Cochin, Paris, France
| | - Magali Rancez
- Laboratory of Dendritic Cells, B Lymphocytes and Cytokines in their Microenvironment During Viral Infections and Cancer, Department of Infection, Immunity and Inflammation, Université de Paris, INSERM, CNRS, Institut Cochin, Paris, France
| |
Collapse
|
7
|
Bruxelle JF, Trattnig N, Mureithi MW, Landais E, Pantophlet R. HIV-1 Entry and Prospects for Protecting against Infection. Microorganisms 2021; 9:microorganisms9020228. [PMID: 33499233 PMCID: PMC7911371 DOI: 10.3390/microorganisms9020228] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 12/19/2022] Open
Abstract
Human Immunodeficiency Virus type-1 (HIV-1) establishes a latent viral reservoir soon after infection, which poses a major challenge for drug treatment and curative strategies. Many efforts are therefore focused on blocking infection. To this end, both viral and host factors relevant to the onset of infection need to be considered. Given that HIV-1 is most often transmitted mucosally, strategies designed to protect against infection need to be effective at mucosal portals of entry. These strategies need to contend also with cell-free and cell-associated transmitted/founder (T/F) virus forms; both can initiate and establish infection. This review will discuss how insight from the current model of HIV-1 mucosal transmission and cell entry has highlighted challenges in developing effective strategies to prevent infection. First, we examine key viral and host factors that play a role in transmission and infection. We then discuss preventive strategies based on antibody-mediated protection, with emphasis on targeting T/F viruses and mucosal immunity. Lastly, we review treatment strategies targeting viral entry, with focus on the most clinically advanced entry inhibitors.
Collapse
Affiliation(s)
- Jean-François Bruxelle
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
- Correspondence: (J.-F.B.); (R.P.)
| | - Nino Trattnig
- Chemical Biology and Drug Discovery, Utrecht University, 3584 CG Utrecht, The Netherlands;
| | - Marianne W. Mureithi
- KAVI—Institute of Clinical Research, College of Health Sciences, University of Nairobi, P.O. Box, Nairobi 19676–00202, Kenya;
| | - Elise Landais
- IAVI Neutralizing Antibody Center, La Jolla, CA 92037, USA;
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Ralph Pantophlet
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
- Correspondence: (J.-F.B.); (R.P.)
| |
Collapse
|
8
|
Liu J, Clayton K, Gao W, Li Y, Zealey C, Budylowski P, Schwartz J, Yue FY, Bie Y, Rini J, Ostrowski M. Trimeric HIV-1 gp140 fused with APRIL, BAFF, and CD40L on the mucosal gp140-specific antibody responses in mice. Vaccine 2020; 38:2149-2159. [PMID: 32014267 DOI: 10.1016/j.vaccine.2020.01.050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 12/04/2019] [Accepted: 01/19/2020] [Indexed: 12/12/2022]
Abstract
HIV-1 envelope (Env)-specific antibody present at mucosal surfaces can block entry of HIV-1 into these portals and thus should be elicited by an HIV-1 preventive vaccine. Since three molecules of tumor necrosis factor superfamily (TNFSF), APRIL, BAFF, and CD40L, could promote mucosal antibody responses, we made fusion constructs of them with an HIV-1 gp140 trimer and tested the mucosal gp140-specific antibody elicited by the fusion constructs in mice using a DNA prime-protein boost vaccination regimen. The fusion constructs formed trimers and displayed both broadly neutralizing antibody epitopes and non-broadly neutralizing antibody epitopes. Compared with the control construct, trimeric gp140, trimeric gp140-APRIL and gp140-BAFF fusion proteins mildly promoted B cell proliferation in vitro, enhanced HIV-1 gp140-binding IgG responses in vaginal lavage or fecal pellets, respectively, and decreased HIV-1 gp140-binding IgA in sera. Gp140-APRIL also augmented HIV-1 gp140-binding IgG in sera. Surprisingly, gp140-CD40L did not promote B cell proliferation in vitro and inhibited mucosal and systemic HIV-1 gp140-binding IgG or IgA. These results suggest that APRIL and BAFF should be further explored as molecular adjuvants for HIV-1 vaccines to enhance mucosal antibody responses, but covalent fusion of TNFSFs to gp140 may hinder their adjuvancy due to steric interactions.
Collapse
Affiliation(s)
- Jun Liu
- Clinical Sciences Division, University of Toronto, Toronto, Ontario, Canada.
| | - Kiera Clayton
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Wenbo Gao
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Yu Li
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Chris Zealey
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Patrick Budylowski
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Jordan Schwartz
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Feng Yun Yue
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Yuan Bie
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - James Rini
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Mario Ostrowski
- Clinical Sciences Division, University of Toronto, Toronto, Ontario, Canada; Department of Immunology, University of Toronto, Toronto, Ontario, Canada; Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
9
|
Aldon Y, Kratochvil S, Shattock RJ, McKay PF. Chemokine-Adjuvanted Plasmid DNA Induces Homing of Antigen-Specific and Non-Antigen-Specific B and T Cells to the Intestinal and Genital Mucosae. THE JOURNAL OF IMMUNOLOGY 2020; 204:903-913. [PMID: 31915263 PMCID: PMC6994839 DOI: 10.4049/jimmunol.1901184] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 11/29/2019] [Indexed: 01/19/2023]
Abstract
Plasmid DNA is a promising vaccine platform that together with electroporation can elicit significant systemic Ab responses; however, immunity at mucosal sites remains low. In this study, we sought to program T and B cells to home to the gastrointestinal and vaginal mucosae using genetic chemokine adjuvants and assessed their impact on immune homeostasis in various distinct immune compartments. BALB/c mice were immunized i.m. with plasmid DNA encoding a model Ag HIV-1 Env gp140 and selected chemokines/cytokine and boosted intravaginally with gp140 recombinant protein. Isolated splenocytes, intestinal lymphocytes, and genital lymphocytes as well as serum and intestinal luminal contents were assessed for Ag-specific reactivity. In addition, flow cytometric analysis was performed to determine the impact on immune homeostasis at these sites. Different molecular chemokine/cytokine adjuvants effected significant alterations to the recruitment of B and T cells to the spleen, vaginal and intestinal mucosae, for example CCL25 enhanced splenic and vaginal Ag-specific T cell responses whereas CCL28 increased the levels of specific T cells only in the vaginal mucosa. The levels of Ab could be modulated in the systemic circulation, as well as the vaginal vault and intestinal lumen, with CCL20 playing a central role. Our data demonstrate that the CCL20, CCL25, and CCL28 genetic chemokine adjuvants enhance the vaccine Ag-specific humoral and cellular responses and induce homing to the intestinal and female genital mucosae.
Collapse
Affiliation(s)
- Yoann Aldon
- Department of Medicine, Imperial College London, London W2 1PG, United Kingdom
| | - Sven Kratochvil
- Department of Medicine, Imperial College London, London W2 1PG, United Kingdom
| | - Robin J Shattock
- Department of Medicine, Imperial College London, London W2 1PG, United Kingdom
| | - Paul F McKay
- Department of Medicine, Imperial College London, London W2 1PG, United Kingdom
| |
Collapse
|
10
|
Perciani CT, Sekhon M, Hundal S, Farah B, Ostrowski MA, Anzala AO, McKinnon LR, Jaoko W, MacDonald KS. Live Attenuated Zoster Vaccine Boosts Varicella Zoster Virus (VZV)-Specific Humoral Responses Systemically and at the Cervicovaginal Mucosa of Kenyan VZV-Seropositive Women. J Infect Dis 2019; 218:1210-1218. [PMID: 29800309 PMCID: PMC6129112 DOI: 10.1093/infdis/jiy320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 05/23/2018] [Indexed: 12/30/2022] Open
Abstract
Background Attenuated varicella zoster virus (VZV) is a promising vector for recombinant vaccines. Because human immunodeficiencyvirus (HIV) vaccines are believed to require mucosal immunogenicity, we characterized mucosal VZV-specific humoral immunity following VZVOka vaccination. Methods Adult Kenyan VZV-seropositive women (n = 44) received a single dose of the live zoster VZVOka vaccine. The anamnestic responses to the virus were followed longitudinally in both plasma and mucosal secretions using an in-house glycoprotein enzyme-linked immunosorbent assay and safety and reactogenicity monitored. VZV seroprevalence and baseline responses to the virus were also characterized in our cohorts (n = 288). Results Besides boosting anti-VZV antibody responses systemically, vaccination also boosted anti-VZV immunity in the cervicovaginal mucosa with a 2.9-fold rise in immunoglobulin G (P < .0001) and 1.6-fold rise in immunoglobulin A (IgA) (P = .004) from the time before immunization and 4 weeks postvaccination. Baseline analysis demonstrated high avidity antibodies at the gastrointestinal and genital mucosa of VZV-seropositive women. Measurement of VZV-specific IgA in saliva is a sensitive tool for detecting prior VZV infection. Conclusions VZVOka vaccine was safe and immunogenic in VZV-seropositive adult Kenyan women. We provided compelling evidence of VZV ability to induce genital mucosa immunity. Clinical Trials Registration NCT02514018.
Collapse
Affiliation(s)
- Catia T Perciani
- Department of Immunology, University of Toronto, Ontario, Canada
| | - Manmeet Sekhon
- Department of Immunology, University of Toronto, Ontario, Canada
| | - Sabrina Hundal
- Department of Immunology, University of Toronto, Ontario, Canada
| | - Bashir Farah
- Kenyan AIDS Vaccine Initiative-Institute of Clinical Research, Nairobi, Kenya
| | - Mario A Ostrowski
- Department of Immunology, University of Toronto, Ontario, Canada.,Keenan Research Centre for Biomedical Science of St Michael's Hospital, Toronto, Ontario, Canada
| | - A Omu Anzala
- Kenyan AIDS Vaccine Initiative-Institute of Clinical Research, Nairobi, Kenya.,Department of Medical Microbiology, University of Nairobi, Kenya
| | - Lyle R McKinnon
- Department of Medical Microbiology, University of Nairobi, Kenya.,Department of Medical Microbiology and Infectious Diseases, Rady College of Medicine, University of Manitoba, Winnipeg, Canada.,Centre for the AIDS Programme of Research in South Africa, Durban
| | - Walter Jaoko
- Kenyan AIDS Vaccine Initiative-Institute of Clinical Research, Nairobi, Kenya.,Department of Medical Microbiology, University of Nairobi, Kenya
| | - Kelly S MacDonald
- Department of Immunology, University of Toronto, Ontario, Canada.,Section of Infectious Diseases, Department of Internal Medicine, Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | | |
Collapse
|
11
|
Sutton MS, Ellis-Connell A, Balgeman AJ, Barry G, Weiler AM, Hetzel SJ, Zhou Y, Lau-Kilby AW, Mason RD, Biris KK, Mascola JR, Sullivan NJ, Roederer M, Friedrich TC, O'Connor SL. CD8β Depletion Does Not Prevent Control of Viral Replication or Protection from Challenge in Macaques Chronically Infected with a Live Attenuated Simian Immunodeficiency Virus. J Virol 2019; 93:e00537-19. [PMID: 31092584 PMCID: PMC6639280 DOI: 10.1128/jvi.00537-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 05/11/2019] [Indexed: 11/20/2022] Open
Abstract
We evaluated the contribution of CD8αβ+ T cells to control of live-attenuated simian immunodeficiency virus (LASIV) replication during chronic infection and subsequent protection from pathogenic SIV challenge. Unlike previous reports with a CD8α-specific depleting monoclonal antibody (mAb), the CD8β-specific mAb CD8β255R1 selectively depleted CD8αβ+ T cells without also depleting non-CD8+ T cell populations that express CD8α, such as natural killer (NK) cells and γδ T cells. Following infusion with CD8β255R1, plasma viremia transiently increased coincident with declining peripheral CD8αβ+ T cells. Interestingly, plasma viremia returned to predepletion levels even when peripheral CD8αβ+ T cells did not. Although depletion of CD8αβ+ T cells in the lymph node (LN) was incomplete, frequencies of these cells were 3-fold lower (P = 0.006) in animals that received CD8β255R1 than in those that received control IgG. It is possible that these residual SIV-specific CD8αβ+ T cells may have contributed to suppression of viremia during chronic infection. We also determined whether infusion of CD8β255R1 in the LASIV-vaccinated animals increased their susceptibility to infection following intravenous challenge with pathogenic SIVmac239. We found that 7/8 animals infused with CD8β255R1, and 3/4 animals infused with the control IgG, were resistant to SIVmac239 infection. These results suggest that infusion with CD8β255R1 did not eliminate the protection afforded to LASIV vaccination. This provides a comprehensive description of the impact of CD8β255R1 infusion on the immunological composition in cynomolgus macaques, compared to an isotype-matched control IgG, while showing that the control of LASIV viremia and protection from challenge can occur even after CD8β255R1 administration.IMPORTANCE Studies of SIV-infected macaques that deplete CD8+ T cells in vivo with monoclonal antibodies have provided compelling evidence for their direct antiviral role. These studies utilized CD8α-specific mAbs that target both the major (CD8αβ+) and minor (CD8αα+) populations of CD8+ T cells but additionally deplete non-CD8+ T cell populations that express CD8α, such as NK cells and γδ T cells. In the current study, we administered the CD8β-specific depleting mAb CD8β255R1 to cynomolgus macaques chronically infected with a LASIV to selectively deplete CD8αβ+ T cells without removing CD8αα+ lymphocytes. We evaluated the impact on control of virus replication and protection from pathogenic SIVmac239 challenge. These results underscore the utility of CD8β255R1 for studying the direct contribution of CD8αβ+ T cells in various disease states.
Collapse
Affiliation(s)
- Matthew S Sutton
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Amy Ellis-Connell
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Alexis J Balgeman
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Gabrielle Barry
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Andrea M Weiler
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Scott J Hetzel
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Yan Zhou
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Annie W Lau-Kilby
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Rosemarie D Mason
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Kristin K Biris
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Nancy J Sullivan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Thomas C Friedrich
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Shelby L O'Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
12
|
Ray K, Mengistu M, Orlandi C, Pazgier M, Lewis GK, DeVico AL. Concurrent Exposure of Neutralizing and Non-neutralizing Epitopes on a Single HIV-1 Envelope Structure. Front Immunol 2019; 10:1512. [PMID: 31338095 PMCID: PMC6628914 DOI: 10.3389/fimmu.2019.01512] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 06/17/2019] [Indexed: 01/14/2023] Open
Abstract
The trimeric envelope spikes on the HIV-1 virus surface initiate infection and comprise key targets for antiviral humoral responses. Circulating virions variably present intact envelope spikes, which react with neutralizing antibodies; and altered envelope structures, which bind non-neutralizing antibodies. Once bound, either type of antibody can enable humoral effector mechanisms with the potential to control HIV-1 infection in vivo. However, it is not clear how the presentation of neutralizing vs. non-neutralizing epitopes defines distinct virus populations and/or envelope structures on single particles. Here we used single-virion fluorescence correlation spectroscopy (FCS), fluorescence resonance energy transfer (FRET), and two-color coincidence FCS approaches to examine whether neutralizing and non-neutralizing antibodies are presented by the same envelope structure. Given the spatial requirements for donor-acceptor energy transfer (≤10 nm), FRET signals generated by paired neutralizing and non-neutralizing fluorescent Fabs should occur via proximal binding to the same target antigen. Fluorescent-labeled Fabs of the neutralizing anti-gp120 antibodies 2G12 and b12 were combined with Fabs of the non-neutralizing anti-gp41 antibody F240, previously thought to mainly bind gp41 "stumps." We find that both 2G12-F240 and/or b12-F240 Fab combinations generate FRET signals on multiple types of virions in solution. FRET efficiencies position the neutralizing and non-neutralizing epitopes between 7.1 and 7.8 nm apart; potentially fitting within the spatial dimensions of a single trimer-derived structure. Further, the frequency of FRET detection suggests that at least one of such structures occurs on the majority of particles in a virus population. Thus, there is frequent, overlapping presentation of non-neutralizing and neutralizing epitope on freely circulating HIV-1 surfaces. Such information provides a broader perspective of how anti-HIV humoral immunity interfaces with circulating virions.
Collapse
Affiliation(s)
- Krishanu Ray
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States,Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, United States,*Correspondence: Krishanu Ray
| | - Meron Mengistu
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States,Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Chiara Orlandi
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States,Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Marzena Pazgier
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States,Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - George K. Lewis
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States,Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Anthony L. DeVico
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States,Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
13
|
Oh JE, Iijima N, Song E, Lu P, Klein J, Jiang R, Kleinstein SH, Iwasaki A. Migrant memory B cells secrete luminal antibody in the vagina. Nature 2019; 571:122-126. [PMID: 31189952 PMCID: PMC6609483 DOI: 10.1038/s41586-019-1285-1] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 05/14/2019] [Indexed: 12/28/2022]
Abstract
Antibodies secreted into mucosal barriers serve to protect the host from a variety of pathogens, and are the basis for successful vaccines1. In type I mucosa (such as the intestinal tract), dimeric IgA secreted by local plasma cells is transported through polymeric immunoglobulin receptors2 and mediates robust protection against viruses3,4. However, owing to the paucity of polymeric immunoglobulin receptors and plasma cells, how and whether antibodies are delivered to the type II mucosa represented by the lumen of the lower female reproductive tract remains unclear. Here, using genital herpes infection in mice, we show that primary infection does not establish plasma cells in the lamina propria of the female reproductive tract. Instead, upon secondary challenge with herpes simplex virus 2, circulating memory B cells that enter the female reproductive tract serve as the source of rapid and robust antibody secretion into the lumen of this tract. CD4 tissue-resident memory T cells secrete interferon-γ, which induces expression of chemokines, including CXCL9 and CXCL10. Circulating memory B cells are recruited to the vaginal mucosa in a CXCR3-dependent manner, and secrete virus-specific IgG2b, IgG2c and IgA into the lumen. These results reveal that circulating memory B cells act as a rapidly inducible source of mucosal antibodies in the female reproductive tract.
Collapse
Affiliation(s)
- Ji Eun Oh
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Norifumi Iijima
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Laboratory of Adjuvant Innovation, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki City, Japan
- Immunology Frontier Research Center, Osaka University, Ibaraki City, Japan
| | - Eric Song
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Peiwen Lu
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Jonathan Klein
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Ruoyi Jiang
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Steven H Kleinstein
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Akiko Iwasaki
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA.
- Department of Molecular Cellular and Developmental Biology, Yale University, New Haven, CT, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
14
|
Abstract
Antibody/antigen binding results in immune complexes (IC) that have a variety of regulatory functions. One important feature is the enhanced host immune activation against antigen contained in the complex. ICs play important roles at several critical steps that lead to B and T cell activation, including antigen targeting/retention, facilitated antigen uptake, antigen presenting cell activation and proper balancing of positive and negative stimulatory signals. In both poultry industry and clinical health care, ICs have been used as preventive and therapeutic vaccines. With our deepening understanding of antibody biology, particularly in light of new revelations of regulatory functions of Fc receptors, mechanistically more precise engineering has spearheaded tailored use of this tool for infection control and cancer therapy. IC-based treatment and prophylaxis have been tested to different extents in HBV, HIV and influenza viral infection control and are actively examined as an alternative treatment for several forms of tumor. As a part of this book series, this chapter aims to discuss the mechanistic aspects of IC signaling and their impact on immune cells. We give samples how this old technology has been used by practitioners over the last several decades and suggest potential paths for future development of IC-based immune therapy.
Collapse
Affiliation(s)
- Yu-Mei Wen
- Key Laboratory of Molecular Virology, Shanghai Medical College, School of Basic Medical Sciences, Fudan University, Shanghai, China. .,Shanghai Medical College, Fudan University, Rm 401, Fuxing Bldg, 131 Yi Xue Yuan Rd, Shanghai, 200032, China.
| | - Yan Shi
- Department of Basic Medical Sciences, Center for Life Sciences, Institute of Immunology, Tsinghua University, Beijing, China.,Department of Microbiology, Immunology & Infectious Diseases and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada.,D301 Medical Sciences Bldg, Tsinghua University, Beijing, 00084, China
| |
Collapse
|
15
|
Kozlowski PA, Aldovini A. Mucosal Vaccine Approaches for Prevention of HIV and SIV Transmission. CURRENT IMMUNOLOGY REVIEWS 2019; 15:102-122. [PMID: 31452652 PMCID: PMC6709706 DOI: 10.2174/1573395514666180605092054] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 04/19/2018] [Accepted: 05/30/2018] [Indexed: 02/06/2023]
Abstract
Optimal protective immunity to HIV will likely require that plasma cells, memory B cells and memory T cells be stationed in mucosal tissues at portals of viral entry. Mucosal vaccine administration is more effective than parenteral vaccine delivery for this purpose. The challenge has been to achieve efficient vaccine uptake at mucosal surfaces, and to identify safe and effective adjuvants, especially for mucosally administered HIV envelope protein immunogens. Here, we discuss strategies used to deliver potential HIV vaccine candidates in the intestine, respiratory tract, and male and female genital tract of humans and nonhuman primates. We also review mucosal adjuvants, including Toll-like receptor agonists, which may adjuvant both mucosal humoral and cellular immune responses to HIV protein immunogens.
Collapse
Affiliation(s)
- Pamela A. Kozlowski
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Anna Aldovini
- Department of Medicine, and Harvard Medical School, Boston Children’s Hospital, Department of Pediatrics, Boston MA, 02115, USA
| |
Collapse
|
16
|
Shang L, Smith AJ, Duan L, Perkey KE, Wietgrefe S, Zupancic M, Southern PJ, Johnson RP, Carlis JV, Haase AT. Vaccine-Associated Maintenance of Epithelial Integrity Correlated With Protection Against Virus Entry. J Infect Dis 2018; 218:1272-1283. [PMID: 29401315 PMCID: PMC6455945 DOI: 10.1093/infdis/jiy062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 01/29/2018] [Indexed: 12/13/2022] Open
Abstract
To identify the mechanisms by which human immunodeficiency virus type 1 (HIV-1) might penetrate the epithelial barrier during sexual transmission to women and the mechanisms of vaccine-associated protection against entry, we characterized early epithelial responses to vaginal inoculation of simian immunodeficiency virus strain mac251 (SIVmac251) in naive or SIVmac239Δnef-vaccinated rhesus macaques. Vaginal inoculation induced an early stress response in the cervicovaginal epithelium, which was associated with impaired epithelial integrity, damaged barrier function, and virus and bacterial translocation. In vaccinated animals, early stress responses were suppressed, and the maintenance of epithelial barrier integrity correlated with prevention of virus entry. These vaccine-protective effects were associated with a previously described mucosal system for locally producing and concentrating trimeric gp41 antibodies at the mucosal interface and with formation of SIV-specific immune complexes that block the stress responses via binding to the epithelial receptor FCGR2B and subsequent inhibitory signaling. Thus, blocking virus entry may be one protective mechanism by which locally concentrated non-neutralizing Ab might prevent HIV sexual transmission to women.
Collapse
Affiliation(s)
- L Shang
- Department of Microbiology and Immunology, Medical School, Minneapolis
| | - A J Smith
- Department of Microbiology and Immunology, Medical School, Minneapolis
| | - L Duan
- Department of Microbiology and Immunology, Medical School, Minneapolis
| | - K E Perkey
- Department of Microbiology and Immunology, Medical School, Minneapolis
| | - S Wietgrefe
- Department of Microbiology and Immunology, Medical School, Minneapolis
| | - M Zupancic
- Department of Microbiology and Immunology, Medical School, Minneapolis
| | - P J Southern
- Department of Microbiology and Immunology, Medical School, Minneapolis
| | - R P Johnson
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia
| | - J V Carlis
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis
| | - A T Haase
- Department of Microbiology and Immunology, Medical School, Minneapolis
| |
Collapse
|
17
|
Brocca-Cofano E, Xu C, Wetzel KS, Cottrell ML, Policicchio BB, Raehtz KD, Ma D, Dunsmore T, Haret-Richter GS, Musaitif K, Keele BF, Kashuba AD, Collman RG, Pandrea I, Apetrei C. Marginal Effects of Systemic CCR5 Blockade with Maraviroc on Oral Simian Immunodeficiency Virus Transmission to Infant Macaques. J Virol 2018; 92:e00576-18. [PMID: 29925666 PMCID: PMC6096825 DOI: 10.1128/jvi.00576-18] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 06/14/2018] [Indexed: 12/20/2022] Open
Abstract
Current approaches do not eliminate all human immunodeficiency virus type 1 (HIV-1) maternal-to-infant transmissions (MTIT); new prevention paradigms might help avert new infections. We administered maraviroc (MVC) to rhesus macaques (RMs) to block CCR5-mediated entry, followed by repeated oral exposure of a CCR5-dependent clone of simian immunodeficiency virus (SIV) mac251 (SIVmac766). MVC significantly blocked the CCR5 coreceptor in peripheral blood mononuclear cells and tissue cells. All control animals and 60% of MVC-treated infant RMs became infected by the 6th challenge, with no significant difference between the number of exposures (P = 0.15). At the time of viral exposures, MVC plasma and tissue (including tonsil) concentrations were within the range seen in humans receiving MVC as a therapeutic. Both treated and control RMs were infected with only a single transmitted/founder variant, consistent with the dose of virus typical of HIV-1 infection. The uninfected RMs expressed the lowest levels of CCR5 on the CD4+ T cells. Ramp-up viremia was significantly delayed (P = 0.05) in the MVC-treated RMs, yet peak and postpeak viral loads were similar in treated and control RMs. In conclusion, in spite of apparent effective CCR5 blockade in infant RMs, MVC had a marginal impact on acquisition and only a minimal impact on the postinfection delay of viremia following oral SIV infection. Newly developed, more effective CCR5 blockers may have a more dramatic impact on oral SIV transmission than MVC.IMPORTANCE We have previously suggested that the very low levels of simian immunodeficiency virus (SIV) maternal-to-infant transmissions (MTIT) in African nonhuman primates that are natural hosts of SIVs are due to a low availability of target cells (CCR5+ CD4+ T cells) in the oral mucosa of the infants, rather than maternal and milk factors. To confirm this new MTIT paradigm, we performed a proof-of-concept study in which we therapeutically blocked CCR5 with maraviroc (MVC) and orally exposed MVC-treated and naive infant rhesus macaques to SIV. MVC had only a marginal effect on oral SIV transmission. However, the observation that the infant RMs that remained uninfected at the completion of the study, after 6 repeated viral challenges, had the lowest CCR5 expression on the CD4+ T cells prior to the MVC treatment appears to confirm our hypothesis, also suggesting that the partial effect of MVC is due to a limited efficacy of the drug. New, more effective CCR5 inhibitors may have a better effect in preventing SIV and HIV transmission.
Collapse
Affiliation(s)
- Egidio Brocca-Cofano
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Cuiling Xu
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Katherine S Wetzel
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mackenzie L Cottrell
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Benjamin B Policicchio
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Infectious Diseases, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kevin D Raehtz
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Dongzhu Ma
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Tammy Dunsmore
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - George S Haret-Richter
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Karam Musaitif
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory, Frederick, Maryland, USA
| | - Brandon F Keele
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory, Frederick, Maryland, USA
| | - Angela D Kashuba
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Ronald G Collman
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ivona Pandrea
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Infectious Diseases, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Cristian Apetrei
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Infectious Diseases, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
18
|
Shang L, Smith AJ, Reilly CS, Duan L, Perkey KE, Wietgrefe S, Zupancic M, Southern PJ, Johnson RP, Carlis JV, Haase AT. Vaccine-modified NF-kB and GR signaling in cervicovaginal epithelium correlates with protection. Mucosal Immunol 2018; 11:512-522. [PMID: 28792003 PMCID: PMC5807226 DOI: 10.1038/mi.2017.69] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 06/19/2017] [Indexed: 02/04/2023]
Abstract
Cervicovaginal epithelium plays a critical role in determining the outcome of virus transmission in the female reproductive tract (FRT) by initiating or suppressing transmission-facilitating mucosal immune responses in naïve and SIVmac239Δnef-vaccinated animals, respectively. In this study, we examined the very early responses of cervical epithelium within 24 h after vaginal exposure to SIV in naive and SIVmac239Δnef-vaccinated rhesus macaques. Using both ex vivo and in vivo experimental systems, we found that vaginal exposure to SIV rapidly induces a broad spectrum of pro-inflammatory responses in the epithelium associated with a reciprocal regulation of NF-kB and glucocorticoid receptor (GR) signaling pathways. Conversely, maintenance of high-level GR expression and suppression of NF-kB expression in the epithelium were associated with an immunologically quiescent state in the FRT mucosa and protection against vaginal challenge in SIVmac239Δnef-vaccinated animals. We show that the immunologically quiescent state is induced by FCGR2B-immune complexes interactions that modify the reciprocal regulation of NF-kB and GR signaling pathways. Our results suggest that targeting the balance of NF-kB and GR signaling in early cervicovaginal epithelium responses could moderate mucosal inflammation and target cell availability after vaginal infection, thereby providing a complementary approach to current prevention strategies.
Collapse
Affiliation(s)
- L Shang
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - A J Smith
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - C S Reilly
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - L Duan
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - K E Perkey
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - S Wietgrefe
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - M Zupancic
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - P J Southern
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - R P Johnson
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - J V Carlis
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - A T Haase
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
19
|
Abstract
Since the discovery of acquired immunodeficiency syndrome (AIDS) in 1981, it has been extremely difficult to develop an effective vaccine or a therapeutic cure despite over 36 years of global efforts. One of the major reasons is due to the lack of an immune-competent animal model that supports live human immunodeficiency virus (HIV) infection and disease progression such that vaccine-induced correlates of protection and efficacy can be determined clearly before human trials. Nevertheless, rhesus macaques infected with simian immunodeficiency virus (SIV) and chimeric simian human immunodeficiency virus (SHIV) have served as invaluable models not only for understanding AIDS pathogenesis but also for studying HIV vaccine and cure. In this chapter, therefore, we summarize major scientific evidence generated in these models since the beginning of the AIDS pandemic. Hopefully, the accumulated knowledge and lessons contributed by thousands of scientists will be useful in promoting the search of an ultimate solution to end HIV/AIDS.
Collapse
|
20
|
Sanders-Beer BE, Voronin Y, McDonald D, Singh A. Harnessing Novel Imaging Approaches to Guide HIV Prevention and Cure Discoveries-A National Institutes of Health and Global HIV Vaccine Enterprise 2017 Meeting Report. AIDS Res Hum Retroviruses 2018; 34:12-26. [PMID: 29145733 DOI: 10.1089/aid.2017.0216] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Advances in imaging technologies have greatly increased our understanding of cellular and molecular interactions in humans and their corresponding animal models of infectious diseases. In the HIV/SIV field, imaging has provided key insights into mucosal viral transmission, local and systemic virus spread, host-virus dynamics, and chronic inflammation/immune activation and the resultant immunopathology. Recent developments in imaging applications are yielding physical, spatial, and temporal measurements to enhance insight into biological functions and disease processes, while retaining important cellular, microenvironmental, organ, and intact organism contextual details. Taking advantage of the latest advancements in imaging technologies may help answer important questions in the HIV field. The Global HIV Vaccine Enterprise in collaboration with the National Institutes of Health (NIH) sponsored a meeting on May 8 and 9, 2017 to provide a platform to review state-of-the-art imaging technologies and to foster multidisciplinary collaborations in HIV/AIDS research. The meeting covered applications of imaging in studies of early events and pathogenesis, reservoirs, and cure, as well as in vaccine development. In addition, presentations and discussions of imaging applications from non-HIV biomedical research areas were included. This report summarizes the presentations and discussions at the meeting.
Collapse
Affiliation(s)
- Brigitte E. Sanders-Beer
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | | | - David McDonald
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Anjali Singh
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
21
|
Pegu A, Hessell AJ, Mascola JR, Haigwood NL. Use of broadly neutralizing antibodies for HIV-1 prevention. Immunol Rev 2017; 275:296-312. [PMID: 28133803 DOI: 10.1111/imr.12511] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Antibodies have a long history in antiviral therapy, but until recently, they have not been actively pursued for HIV-1 due to modest potency and breadth of early human monoclonal antibodies (MAbs) and perceived insurmountable technical, financial, and logistical hurdles. Recent advances in the identification and characterization of MAbs with the ability to potently neutralize diverse HIV-1 isolates have reinvigorated discussion and testing of these products in humans, since new broadly neutralizing MAbs (bnMAbs) are more likely to be effective against worldwide strains of HIV-1. In animal models, there is abundant evidence that bnMAbs can block infection in a dose-dependent manner, and the more potent bnMAbs will allow clinical testing at infusion doses that are practically achievable. Moreover, recent advances in antibody engineering are providing further improvements in MAb potency, breadth, and half-life. This review summarizes the current state of the field of bnMAb protection in animal models as well as a review of variables that are critical for antiviral activity. Several bnMAbs are currently in clinical testing, and we offer perspectives on their use as pre-exposure prophylaxis (PrEP), potential benefits beyond sterilizing immunity, and a discussion of future approaches to engineer novel molecules.
Collapse
Affiliation(s)
| | - Ann J Hessell
- Oregon National Primate Center, Oregon Health & Science University, Beaverton, OR, USA
| | | | - Nancy L Haigwood
- Oregon National Primate Center, Oregon Health & Science University, Beaverton, OR, USA
| |
Collapse
|
22
|
Buchbinder SP, Grunenberg NA, Sanchez BJ, Seaton KE, Ferrari G, Moody MA, Frahm N, Montefiori DC, Hay CM, Goepfert PA, Baden LR, Robinson HL, Yu X, Gilbert PB, McElrath MJ, Huang Y, Tomaras GD, on behalf of the HIV Vaccine Trials Network (HVTN) 094 Study Group. Immunogenicity of a novel Clade B HIV-1 vaccine combination: Results of phase 1 randomized placebo controlled trial of an HIV-1 GM-CSF-expressing DNA prime with a modified vaccinia Ankara vaccine boost in healthy HIV-1 uninfected adults. PLoS One 2017; 12:e0179597. [PMID: 28727817 PMCID: PMC5519050 DOI: 10.1371/journal.pone.0179597] [Citation(s) in RCA: 25] [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: 02/19/2017] [Accepted: 05/30/2017] [Indexed: 12/20/2022] Open
Abstract
Background A phase 1 trial of a clade B HIV vaccine in HIV-uninfected adults evaluated the safety and immunogenicity of a DNA prime co-expressing GM-CSF (Dg) followed by different numbers and intervals of modified vaccinia Ankara Boosts (M). Both vaccines produce virus-like particles presenting membrane-bound Env. Methods Four US sites randomized 48 participants to receiving 1/10th the DNA dose as DgDgMMM given at 0, 2, 4, 6 and 8 months, or full dose DgDgM_M or DgDgMM_M regimens, given at 0, 2, 4, and 8 months, and 0, 2, 4, 6, and 10 months, respectively. Peak immunogenicity was measured 2 weeks post-last vaccination. Results All regimens were well tolerated and safe. Full dose DgDgM_M and DgDgMM_M regimens generated Env-specific IgG to HIV-1 Env in >90%, IgG3 in >80%, and IgA in <20% of participants. Responses to gp140 and gp41 targets were more common and of higher magnitude than to gp120 and V1V2. The gp41 antibody included reactivity to the conserved immunodominant region with specificities known to mediate virus capture and phagocytosis and did not cross-react with a panel of intestinal flora antigens. The 3rd dose of MVA increased the avidity of elicited antibody (7.5% to 39%), the ADCC response to Bal gp120 (14% to 64%), and the one-year durability of the IgG3 responses to gp41 by 4-fold (13% vs. 3.5% retention of peak response). The co-expressed GM-CSF did not enhance responses over those in trials testing this vaccine without GM-CSF. Conclusion This DNA/MVA prime-boost regimen induced durable, functional humoral responses that included ADCC, high antibody avidity, and Env IgG1 and IgG3 binding responses to the immunodominant region of gp41. The third, spaced MVA boost improved the overall quality of the antibody response. These products without co-expressed GM-CSF but combined with protein boosts will be considered for efficacy evaluation. Trial registration ClinicalTrials.gov NCT01571960
Collapse
Affiliation(s)
- Susan P. Buchbinder
- Bridge HIV, San Francisco Department of Public Health, San Francisco, California, United States of America
- Departments of Medicine, Epidemiology and Biostatistics, University of California, San Francisco, California, United States of America
- * E-mail:
| | - Nicole A. Grunenberg
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Brittany J. Sanchez
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Kelly E. Seaton
- Department of Surgery, Duke Human Vaccine Institute, Durham, North Carolina, United States of America
| | - Guido Ferrari
- Department of Surgery, Duke Human Vaccine Institute, Durham, North Carolina, United States of America
| | - M. Anthony Moody
- Department of Surgery, Duke Human Vaccine Institute, Durham, North Carolina, United States of America
| | - Nicole Frahm
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - David C. Montefiori
- Department of Surgery, Duke Human Vaccine Institute, Durham, North Carolina, United States of America
| | - Christine M. Hay
- Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Paul A. Goepfert
- Department of Medicine, University of Alabama, Birmingham, Alabama, United States of America
| | - Lindsey R. Baden
- Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | | | - Xuesong Yu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Peter B. Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Biostatistics, 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
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Yunda Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Georgia D. Tomaras
- Department of Surgery, Duke Human Vaccine Institute, Durham, North Carolina, United States of America
| | | |
Collapse
|
23
|
Brandenberg OF, Magnus C, Rusert P, Günthard HF, Regoes RR, Trkola A. Predicting HIV-1 transmission and antibody neutralization efficacy in vivo from stoichiometric parameters. PLoS Pathog 2017; 13:e1006313. [PMID: 28472201 PMCID: PMC5417720 DOI: 10.1371/journal.ppat.1006313] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 03/24/2017] [Indexed: 01/08/2023] Open
Abstract
The potential of broadly neutralizing antibodies targeting the HIV-1 envelope trimer to prevent HIV-1 transmission has opened new avenues for therapies and vaccines. However, their implementation remains challenging and would profit from a deepened mechanistic understanding of HIV-antibody interactions and the mucosal transmission process. In this study we experimentally determined stoichiometric parameters of the HIV-1 trimer-antibody interaction, confirming that binding of one antibody is sufficient for trimer neutralization. This defines numerical requirements for HIV-1 virion neutralization and thereby enables mathematical modelling of in vitro and in vivo antibody neutralization efficacy. The model we developed accurately predicts antibody efficacy in animal passive immunization studies and provides estimates for protective mucosal antibody concentrations. Furthermore, we derive estimates of the probability for a single virion to start host infection and the risks of male-to-female HIV-1 transmission per sexual intercourse. Our work thereby delivers comprehensive quantitative insights into both the molecular principles governing HIV-antibody interactions and the initial steps of mucosal HIV-1 transmission. These insights, alongside the underlying, adaptable modelling framework presented here, will be valuable for supporting in silico pre-trial planning and post-hoc evaluation of HIV-1 vaccination or antibody treatment trials.
Collapse
Affiliation(s)
| | - Carsten Magnus
- Institute of Medical Virology, University of Zürich, Zurich, Switzerland
| | - Peter Rusert
- Institute of Medical Virology, University of Zürich, Zurich, Switzerland
| | - Huldrych F. Günthard
- Institute of Medical Virology, University of Zürich, Zurich, Switzerland
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
| | - Roland R. Regoes
- Institute of Integrative Biology, ETH Zurich, Zurich, Switzerland
| | - Alexandra Trkola
- Institute of Medical Virology, University of Zürich, Zurich, Switzerland
- * E-mail:
| |
Collapse
|
24
|
Distinct transcriptome profiles of Gag-specific CD8+ T cells temporally correlated with the protection elicited by SIVΔnef live attenuated vaccine. PLoS One 2017; 12:e0173929. [PMID: 28333940 PMCID: PMC5363825 DOI: 10.1371/journal.pone.0173929] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 02/27/2017] [Indexed: 12/21/2022] Open
Abstract
The live attenuated vaccine (LAV) SIVmac239Δnef (SIVΔnef) confers the best protection among all the vaccine modalities tested in rhesus macaque model of HIV-1 infection. This vaccine has a unique feature of time-dependent protection: macaques are not protected at 3–5 weeks post vaccination (WPV), whereas immune protection emerges between 15 and 20 WPV. Although the exact mechanisms of the time-dependent protection remain incompletely understood, studies suggested that both cellular and humoral immunities contribute to this time-dependent protection. To further elucidate the mechanisms of protection induced by SIVΔnef, we longitudinally compared the global gene expression profiles of SIV Gag-CM9+ CD8+ (Gag-specific CD8+) T cells from peripheral blood of Mamu-A*01+ rhesus macaques at 3 and 20 WPV using rhesus microarray. We found that gene expression profiles of Gag-specific CD8+ T cells at 20 WPV are qualitatively different from those at 3 WPV. At 20 WPV, the most significant transcriptional changes of Gag-specific CD8+ T cells were genes involved in TCR signaling, differentiation and maturation toward central memory cells, with increased expression of CCR7, TCRα, TCRβ, CD28 and decreased expression of CTLA-4, IFN-γ, RANTES, granzyme A and B. Our study suggests that a higher quality of SIV-specific CD8+ T cells elicited by SIVΔnef over time contributes to the maturation of time-dependent protection.
Collapse
|
25
|
Wang N, Yuan Z, Niu W, Li Q, Guo J. Synthetic biology approach for the development of conditionally replicating HIV-1 vaccine. JOURNAL OF CHEMICAL TECHNOLOGY AND BIOTECHNOLOGY (OXFORD, OXFORDSHIRE : 1986) 2017; 92:455-462. [PMID: 28983143 PMCID: PMC5624719 DOI: 10.1002/jctb.5174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
While the combined antiretroviral therapy has resulted in a significant decrease in HIV-1 related morbidity and mortality, the HIV-1 pandemic has not been substantially averted. To curtail the 2.4 million new infections each year, a prophylactic HIV-1 vaccine is urgently needed. This review first summarizes four major completed clinical efficacy trials of prophylactic HIV-1 vaccine and their outcomes. Next, it discusses several other approaches that have not yet advanced to clinical efficacy trials, but provided valuable insights into vaccine design. Among them, live-attenuated vaccines (LAVs) provided excellent protection in a non-human primate model. However, safety concerns have precluded the current version of LAVs from clinical application. As the major component of this review, two synthetic biology approaches for improving the safety of HIV-1 LAVs through controlling HIV-1 replication are discussed. Particular focus is on a novel approach that uses unnatural amino acid-mediated suppression of amber nonsense codon to generate conditionally replicating HIV-1 variants. The objective is to attract more attention towards this promising research field and to provoke creative designs and innovative utilization of the two control strategies.
Collapse
Affiliation(s)
- Nanxi Wang
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, 68588, USA
| | - Zhe Yuan
- Nebraska Center for Virology & School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, 68588, USA
| | - Wei Niu
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska, 68588, USA
| | - Qingsheng Li
- Nebraska Center for Virology & School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, 68588, USA
| | - Jiantao Guo
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, 68588, USA
| |
Collapse
|
26
|
Shang L, Duan L, Perkey KE, Wietgrefe S, Zupancic M, Smith AJ, Southern PJ, Johnson RP, Haase AT. Epithelium-innate immune cell axis in mucosal responses to SIV. Mucosal Immunol 2017; 10:508-519. [PMID: 27435105 PMCID: PMC5250613 DOI: 10.1038/mi.2016.62] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 06/18/2016] [Indexed: 02/04/2023]
Abstract
In the SIV (simian immunodeficiency virus)-rhesus macaque model of HIV-1 (human immunodeficiency virus type I) transmission to women, one hallmark of the mucosal response to exposure to high doses of SIV is CD4 T-cell recruitment that fuels local virus expansion in early infection. In this study, we systematically analyzed the cellular events and chemoattractant profiles in cervical tissues that precede CD4 T-cell recruitment. We show that vaginal exposure to the SIV inoculum rapidly induces chemokine expression in cervical epithelium including CCL3, CCL20, and CXCL8. The chemokine expression is associated with early recruitment of macrophages and plasmacytoid dendritic cells that are co-clustered underneath the cervical epithelium. Production of chemokines CCL3 and CXCL8 by these cells in turn generates a chemokine gradient that is spatially correlated with the recruitment of CD4 T cells. We further show that the protection of SIVmac239Δnef vaccination against vaginal challenge is correlated with the absence of this epithelium-innate immune cell-CD4 T-cell axis response in the cervical mucosa. Our results reveal a critical role for cervical epithelium in initiating early mucosal responses to vaginal infection, highlight an important role for macrophages in target cell recruitment, and provide further evidence of a paradoxical dampening effect of a protective vaccine on these early mucosal responses.
Collapse
Affiliation(s)
- L Shang
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - L Duan
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - K E Perkey
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - S Wietgrefe
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - M Zupancic
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - A J Smith
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - P J Southern
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - R P Johnson
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - A T Haase
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
27
|
Boesch AW, Miles AR, Chan YN, Osei-Owusu NY, Ackerman ME. IgG Fc variant cross-reactivity between human and rhesus macaque FcγRs. MAbs 2017; 9:455-465. [PMID: 28055295 DOI: 10.1080/19420862.2016.1274845] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Non-human primate (NHP) studies are often an essential component of antibody development efforts before human trials. Because the efficacy or toxicity of candidate antibodies may depend on their interactions with Fcγ receptors (FcγR) and their resulting ability to induce FcγR-mediated effector functions such as antibody-dependent cell-meditated cytotoxicity and phagocytosis (ADCP), the evaluation of human IgG variants with modulated affinity toward human FcγR is becoming more prevalent in both infectious disease and oncology studies in NHP. Reliable translation of these results necessitates analysis of the cross-reactivity of these human Fc variants with NHP FcγR. We report evaluation of the binding affinities of a panel of human IgG subclasses, Fc amino acid point mutants and Fc glycosylation variants against the common allotypes of human and rhesus macaque FcγR by applying a high-throughput array-based surface plasmon resonance platform. The resulting data indicate that amino acid variation present in rhesus FcγRs can result in disrupted, matched, or even increased affinity of IgG Fc variants compared with human FcγR orthologs. These observations emphasize the importance of evaluating species cross-reactivity and developing an understanding of the potential limitations or suitability of representative in vitro and in vivo models before human clinical studies when either efficacy or toxicity may be associated with FcγR engagement.
Collapse
Affiliation(s)
- Austin W Boesch
- a Thayer School of Engineering, Dartmouth College , Hanover , NH , USA
| | - Adam R Miles
- b Wasatch Microfluidics , Salt Lake City , UT , USA
| | - Ying N Chan
- a Thayer School of Engineering, Dartmouth College , Hanover , NH , USA
| | - Nana Y Osei-Owusu
- c Department of Microbiology and Immunology , Geisel School of Medicine , Lebanon , NH , USA
| | - Margaret E Ackerman
- a Thayer School of Engineering, Dartmouth College , Hanover , NH , USA.,c Department of Microbiology and Immunology , Geisel School of Medicine , Lebanon , NH , USA
| |
Collapse
|
28
|
Abstract
It is clear that antibodies can play a pivotal role in preventing the transmission of HIV-1 and large efforts to identify an effective antibody-based vaccine to quell the epidemic. Shortly after HIV-1 was discovered as the cause of AIDS, the search for epitopes recognized by neutralizing antibodies became the driving strategy for an antibody-based vaccine. Neutralization escape variants were discovered shortly thereafter, and, after almost three decades of investigation, it is now known that autologous neutralizing antibody responses and their selection of neutralization resistant HIV-1 variants can lead to broadly neutralizing antibodies in some infected individuals. This observation drives an intensive effort to identify a vaccine to elicit broadly neutralizing antibodies. In contrast, there has been less systematic study of antibody specificities that must rely mainly or exclusively on other protective mechanisms, although non-human primate (NHP) studies as well as the RV144 vaccine trial indicate that non-neutralizing antibodies can contribute to protection. Here we propose a novel strategy to identify new epitope targets recognized by these antibodies for which viral escape is unlikely or impossible.
Collapse
Affiliation(s)
- George K Lewis
- Division of Vaccine Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Marzena Pazgier
- Division of Vaccine Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Anthony L DeVico
- Division of Vaccine Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
29
|
Abstract
The scale and scope of the global epidemic, coupled to challenges with traditional vaccine development approaches, point toward a need for novel methodologies for HIV vaccine research. While the development of vaccines able to induce broadly neutralizing antibodies remains the ultimate goal, to date, vaccines continue to fail to induce these rare humoral immune responses. Conversely, growing evidence across vaccine platforms in both non-human primates and humans points to a role for polyclonal vaccine-induced antibody responses in protection from infection. These candidate vaccines, despite employing disparate viral vectors and immunization strategies, consistently identify a role for functional or non-traditional antibody activities as correlates of immunity. However, the precise mechanism(s) of action of these "binding" antibodies, their specific characteristics, and their ability to be selectively induced and/or potentiated to result in complete protection merits parallel investigation to neutralizing antibody-based vaccine design approaches. Ultimately, while neutralizing and functional antibody-based vaccine strategies need not be mutually exclusive, defining the specific characteristics of "protective" functional antibodies may provide a target immune profile to potentially induce more robust immunity against HIV. Specifically, one approach to guide the development of functional antibody-based vaccine strategies, termed "systems serology", offers an unbiased and comprehensive approach to systematically survey humoral immune responses, capturing the array of functions and humoral response characteristics that may be induced following vaccination with high resolution. Coupled to machine learning tools, large datasets that explore the "antibody-ome" offer a means to step back from anticipated correlates and mechanisms of protection and toward a more fundamental understanding of coordinated aspects of humoral immune responses, to more globally differentiate among vaccine candidates, and most critically, to identify the features of humoral immunity that distinguish protective from non-protective responses. Overall, the systematic serological approach described here aimed at broadly capturing the enormous biodiversity in antibody profiles that may emerge following vaccination, complements the existing cutting edge tools in the cellular immunology space that survey vaccine-induced polyfunctional cellular activity by flow cytometry, transcriptional profiling, epigenetic, and metabolomic analysis to offer a means to develop both a more nuanced and a more complete understanding of correlates of protection to support the design of functional vaccine strategies.
Collapse
Affiliation(s)
| | - Dan H Barouch
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Beth Israel Deaconness Medical Center, Boston, MA, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| |
Collapse
|
30
|
Adnan S, Reeves RK, Gillis J, Wong FE, Yu Y, Camp JV, Li Q, Connole M, Li Y, Piatak M, Lifson JD, Li W, Keele BF, Kozlowski PA, Desrosiers RC, Haase AT, Johnson RP. Persistent Low-Level Replication of SIVΔnef Drives Maturation of Antibody and CD8 T Cell Responses to Induce Protective Immunity against Vaginal SIV Infection. PLoS Pathog 2016; 12:e1006104. [PMID: 27959961 PMCID: PMC5189958 DOI: 10.1371/journal.ppat.1006104] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 12/27/2016] [Accepted: 11/30/2016] [Indexed: 12/31/2022] Open
Abstract
Defining the correlates of immune protection conferred by SIVΔnef, the most effective vaccine against SIV challenge, could enable the design of a protective vaccine against HIV infection. Here we provide a comprehensive assessment of immune responses that protect against SIV infection through detailed analyses of cellular and humoral immune responses in the blood and tissues of rhesus macaques vaccinated with SIVΔnef and then vaginally challenged with wild-type SIV. Despite the presence of robust cellular immune responses, animals at 5 weeks after vaccination displayed only transient viral suppression of challenge virus, whereas all macaques challenged at weeks 20 and 40 post-SIVΔnef vaccination were protected, as defined by either apparent sterile protection or significant suppression of viremia in infected animals. Multiple parameters of CD8 T cell function temporally correlated with maturation of protection, including polyfunctionality, phenotypic differentiation, and redistribution to gut and lymphoid tissues. Importantly, we also demonstrate the induction of a tissue-resident memory population of SIV-specific CD8 T cells in the vaginal mucosa, which was dependent on ongoing low-level antigenic stimulation. Moreover, we show that vaginal and serum antibody titers inversely correlated with post-challenge peak viral load, and we correlate the accumulation and affinity maturation of the antibody response to the duration of the vaccination period as well as to the SIVΔnef antigenic load. In conclusion, maturation of SIVΔnef-induced CD8 T cell and antibody responses, both propelled by viral persistence in the gut mucosa and secondary lymphoid tissues, results in protective immune responses that are able to interrupt viral transmission at mucosal portals of entry as well as potential sites of viral dissemination.
Collapse
Affiliation(s)
- Sama Adnan
- Yerkes National Primate Research Center, Emory University, Atlanta GA, United States of America
- New England Primate Research Center, Harvard Medical School, Southborough Campus, Pine Hill Drive, Southborough, MA, United States of America
| | - R. Keith Reeves
- New England Primate Research Center, Harvard Medical School, Southborough Campus, Pine Hill Drive, Southborough, MA, United States of America
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Jacqueline Gillis
- New England Primate Research Center, Harvard Medical School, Southborough Campus, Pine Hill Drive, Southborough, MA, United States of America
| | - Fay E. Wong
- New England Primate Research Center, Harvard Medical School, Southborough Campus, Pine Hill Drive, Southborough, MA, United States of America
| | - Yi Yu
- New England Primate Research Center, Harvard Medical School, Southborough Campus, Pine Hill Drive, Southborough, MA, United States of America
| | - Jeremy V. Camp
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Qingsheng Li
- Nebraska Center for Virology and School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | - Michelle Connole
- New England Primate Research Center, Harvard Medical School, Southborough Campus, Pine Hill Drive, Southborough, MA, United States of America
| | - Yuan Li
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Michael Piatak
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Wenjun Li
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, United States of America
| | - Brandon F. Keele
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Pamela A. Kozlowski
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Ronald C. Desrosiers
- Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Ashley T. Haase
- Department of Microbiology, Medical School, University of Minnesota, MMC 196, 420 Delaware Street S.E., Minneapolis, Minnesota, United States of America
| | - R. Paul Johnson
- Yerkes National Primate Research Center, Emory University, Atlanta GA, United States of America
- New England Primate Research Center, Harvard Medical School, Southborough Campus, Pine Hill Drive, Southborough, MA, United States of America
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States of America
| |
Collapse
|
31
|
Human vaginal fluid contains exosomes that have an inhibitory effect on an early step of the HIV-1 life cycle. AIDS 2016; 30:2611-2616. [PMID: 27536982 DOI: 10.1097/qad.0000000000001236] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Vaginal transmission is crucial to the spread of HIV-1 around the world. It is not yet clear what type (s) of innate defenses against HIV-1 infection are present in the vagina. Here, we aimed to determine whether human vaginal fluid contains exosomes that may possess anti-HIV-1 activity. METHODS The exosomal fraction was isolated from samples of vaginal fluids. The presence of exosomes was confirmed by flow cytometry and western blotting. The newly discovered exosomes were tested for their ability to block early steps of HIV-1 infection in vitro using established cell culture systems and real time PCR-based methods. RESULTS Vaginal fluid contains exosomes expressing CD9, CD63, and CD81 exosomal markers. The exosomal fraction of the fluid-reduced transmission of HIV-1 vectors by 60%, the efficiency of reverse transcription step by 58.4%, and the efficiency of integration by 47%. Exosomes had no effect on the entry of HIV-1 vectors. CONCLUSION Human vaginal fluid exosomes are newly discovered female innate defenses that may protect women against HIV-1 infection.
Collapse
|
32
|
Reproducing SIVΔnef vaccine correlates of protection: trimeric gp41 antibody concentrated at mucosal front lines. AIDS 2016; 30:2427-2438. [PMID: 27428745 DOI: 10.1097/qad.0000000000001199] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Vaccination with SIVmac239Δnef provides robust protection against subsequent challenge with wild-type simian immunodeficiency virus (SIV), but safety issues have precluded designing an HIV-1 vaccine based on a live-attenuated virus concept. Safe immunogens and adjuvants that could reproduce identified immune correlates of SIVmac239Δnef protection therefore offer an alternative path for development of an HIV vaccine. Here we describe SIV envelope trimeric gp41 (gp41t) immunogens based on a protective correlate of antibodies to gp41t concentrated on the path of virus entry by the neonatal Fc receptor (FcRn) in cervical vaginal epithelium. We developed a gp41t immunogen-monophosphoryl lipid A adjuvant liposomal nanoparticle for intramuscular (i.m.) immunization and a gp41t-Fc immunogen for intranasal immunization for pilot studies in mice, rabbits, and rhesus macaques. Repeated immunizations to mimic persistent antigen exposure in infection elicited gp41t antibodies in rhesus macaques that were detectable in FcRn+ cervical vaginal epithelium, thus recapitulating one key feature of SIVmac239Δnef vaccinated and protected animals. Although this strategy did not reproduce the system of local production of antibody in SIVmac239Δnef-vaccinated animals, passive immunization experiments supported the concept that sufficiently high levels of antibody can be concentrated by the FcRn at mucosal frontlines, thus setting the stage for assessing protection against vaginal challenge by gp41t immunization.
Collapse
|
33
|
Non-neutralizing antibody functions for protection and control HIV in humans and SIV and SHIV in non-human primates. AIDS 2016; 30:2551-2553. [PMID: 27753680 DOI: 10.1097/qad.0000000000001200] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
34
|
Aravantinou M, Frank I, Hallor M, Singer R, Tharinger H, Kenney J, Gettie A, Grasperge B, Blanchard J, Salazar A, Piatak M, Lifson JD, Robbiani M, Derby N. PolyICLC Exerts Pro- and Anti-HIV Effects on the DC-T Cell Milieu In Vitro and In Vivo. PLoS One 2016; 11:e0161730. [PMID: 27603520 PMCID: PMC5014349 DOI: 10.1371/journal.pone.0161730] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 07/14/2016] [Indexed: 12/24/2022] Open
Abstract
Myeloid dendritic cells (mDCs) contribute to both HIV pathogenesis and elicitation of antiviral immunity. Understanding how mDC responses to stimuli shape HIV infection outcomes will inform HIV prevention and treatment strategies. The long double-stranded RNA (dsRNA) viral mimic, polyinosinic polycytidylic acid (polyIC, PIC) potently stimulates DCs to focus Th1 responses, triggers direct antiviral activity in vitro, and boosts anti-HIV responses in vivo. Stabilized polyICLC (PICLC) is being developed for vaccine adjuvant applications in humans, making it critical to understand how mDC sensing of PICLC influences HIV infection. Using the monocyte-derived DC (moDC) model, we sought to describe how PICLC (vs. other dsRNAs) impacts HIV infection within DCs and DC-T cell mixtures. We extended this work to in vivo macaque rectal transmission studies by administering PICLC with or before rectal SIVmac239 (SIVwt) or SIVmac239ΔNef (SIVΔNef) challenge. Like PIC, PICLC activated DCs and T cells, increased expression of α4β7 and CD169, and induced type I IFN responses in vitro. The type of dsRNA and timing of dsRNA exposure differentially impacted in vitro DC-driven HIV infection. Rectal PICLC treatment similarly induced DC and T cell activation and pro- and anti-HIV factors locally and systemically. Importantly, this did not enhance SIV transmission in vivo. Instead, SIV acquisition was marginally reduced after a single high dose challenge. Interestingly, in the PICLC-treated, SIVΔNef-infected animals, SIVΔNef viremia was higher, in line with the importance of DC and T cell activation in SIVΔNef replication. In the right combination anti-HIV strategy, PICLC has the potential to limit HIV infection and boost HIV immunity.
Collapse
Affiliation(s)
- Meropi Aravantinou
- Center for Biomedical Research, Population Council, New York, NY, United States of America
| | - Ines Frank
- Center for Biomedical Research, Population Council, New York, NY, United States of America
| | - Magnus Hallor
- Center for Biomedical Research, Population Council, New York, NY, United States of America
- Linköping University, Linköping, Sweden
| | - Rachel Singer
- Center for Biomedical Research, Population Council, New York, NY, United States of America
| | - Hugo Tharinger
- Center for Biomedical Research, Population Council, New York, NY, United States of America
| | - Jessica Kenney
- Center for Biomedical Research, Population Council, New York, NY, United States of America
| | - Agegnehu Gettie
- Aaron Diamond AIDS Research Center, Rockefeller University, New York, NY, United States of America
| | - Brooke Grasperge
- Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, LA, United States of America
| | - James Blanchard
- Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, LA, United States of America
| | | | - Michael Piatak
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, MD, United States of America
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, MD, United States of America
| | - Melissa Robbiani
- Center for Biomedical Research, Population Council, New York, NY, United States of America
| | - Nina Derby
- Center for Biomedical Research, Population Council, New York, NY, United States of America
| |
Collapse
|
35
|
Lama JR, Karuna ST, Grant SP, Swann EM, Ganoza C, Segura P, Montano SM, Lacherre M, De Rosa SC, Buchbinder S, Sanchez J, McElrath MJ, Lemos MP, HVTN 914 Study Team. Transient Peripheral Immune Activation follows Elective Sigmoidoscopy or Circumcision in a Cohort Study of MSM at Risk of HIV Infection. PLoS One 2016; 11:e0160487. [PMID: 27536938 PMCID: PMC4990246 DOI: 10.1371/journal.pone.0160487] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 07/20/2016] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Rectal and genital sampling in HIV prevention trials permits assessments at the site of HIV entry. Yet the safety and acceptability of circumcision and sigmoidoscopy (and associated abstinence recommendations) are unknown in uncircumcised men who have sex with men (MSM) at high risk of HIV infection. METHODS Twenty-nine HIV-seronegative high-risk Peruvian MSM agreed to elective sigmoidoscopy biopsy collections (weeks 2 and 27) and circumcision (week 4) in a 28-week cohort study designed to mimic an HIV vaccine study mucosal collection protocol. We monitored adherence to abstinence recommendations, procedure-related complications, HIV infections, peripheral immune activation, and retention. RESULTS Twenty-three (79.3%) underwent a first sigmoidoscopy, 21 (72.4%) were circumcised, and 16 (55.2%) completed a second sigmoidoscopy during the study period. All who underwent procedures completed the associated follow-up safety visits. Those completing the procedures reported they were well tolerated, and complication rates were similar to those reported in the literature. Immune activation was detected during the healing period (1 week post-sigmoidoscopy, 6 weeks post-circumcision), including increases in CCR5+CD4+T cells and α4β7+CD4+T cells. Most participants adhered to post-circumcision abstinence recommendations whereas reduced adherence occurred post-sigmoidoscopy. CONCLUSION Rectosigmoid mucosal and genital tissue collections were safe in high-risk MSM. Although the clinical implications of the post-procedure increase in peripheral immune activation markers are unknown, they reinforce the need to provide ongoing risk reduction counseling and support for post-procedure abstinence recommendations. Future HIV vaccine studies should also consider the effects of mucosal and tissue collections on peripheral blood endpoints in trial design and analysis. TRIAL REGISTRATION ClinicalTrials.gov NCT02630082.
Collapse
Affiliation(s)
| | - Shelly T. Karuna
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Shannon P. Grant
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Edith M. Swann
- Vaccine Clinical Research Branch, Division of AIDS, National Institutes of Allergy and Infectious Diseases, US 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
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Susan Buchbinder
- San Francisco Department of Health, San Francisco, California, United States of America
| | - Jorge Sanchez
- Asociacion Civil Impacta Salud y Educacion, Lima, Peru
- Department of Global Health, 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
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Maria P. Lemos
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | | |
Collapse
|
36
|
MOLDT B, LE K, CARNATHAN DG, WHITNEY JB, SCHULTZ N, LEWIS MG, BORDUCCHI E, SMITH K, MACKEL JJ, SWEAT SL, HODGES AP, GODZIK A, PARREN PWHI, SILVESTRI G, BAROUCH DH, BURTON DR. Neutralizing antibody affords comparable protection against vaginal and rectal simian/human immunodeficiency virus challenge in macaques. AIDS 2016; 30:1543-51. [PMID: 27243773 PMCID: PMC4915739 DOI: 10.1097/qad.0000000000001102] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Passive administration of broadly neutralizing antibodies has been shown to protect against both vaginal and rectal challenge in the simian/human immunodeficiency virus (SHIV)/macaque model of HIV transmission. However, the relative efficacy of antibody against the two modes of exposure is unknown and, given differences in the composition and immunology of the two tissue compartments, this is an important gap in knowledge. To investigate the significance of the challenge route for antibody-mediated protection, we performed a comparative protection study in macaques using the highly potent human monoclonal antibody, PGT126. DESIGN Animals were administered PGT126 at three different doses before challenged either vaginally or rectally with a single dose of SHIVSF163P3. METHODS Viral loads, PGT126 serum concentrations, and serum neutralizing titers were monitored. RESULTS In vaginally challenged animals, sterilizing immunity was achieved in all animals administered 10 mg/kg, in two of five animals administered 2 mg/kg and in one of five animals administered 0.4 mg/kg PGT126. Comparable protection was observed for the corresponding groups challenged rectally as sterilizing immunity was achieved in three of four animals administered 10 mg/kg, in two of four animals administered 2 mg/kg and in none of four animals administered 0.4 mg/kg PGT126. Serological analysis showed similar serum concentrations of PGT126 and serum neutralization titers in animals administered the same antibody dose. CONCLUSION Our data suggest that broadly neutralizing antibody-mediated protection is not strongly dependent on the mucosal route of challenge, which indicates that a vaccine aimed to induce a neutralizing antibody response would have broadly similar efficacy against both primary transmission routes for HIV.
Collapse
Affiliation(s)
- Brian MOLDT
- Department of Immunology and Microbial Science, IAVI Neutralizing Antibody Center and Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, California, USA
| | - Khoa LE
- Department of Immunology and Microbial Science, IAVI Neutralizing Antibody Center and Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, California, USA
| | - Diane G. CARNATHAN
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - James B. WHITNEY
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Niccole SCHULTZ
- Department of Immunology and Microbial Science, IAVI Neutralizing Antibody Center and Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, California, USA
| | | | - Erica BORDUCCHI
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Kaitlin SMITH
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Joseph J. MACKEL
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Shelby L. SWEAT
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Andrew P. HODGES
- Bioinformatics and Systems Biology Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Adam GODZIK
- Bioinformatics and Systems Biology Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | | | - Guido SILVESTRI
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Dan H. BAROUCH
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Dennis R. BURTON
- Department of Immunology and Microbial Science, IAVI Neutralizing Antibody Center and Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, California, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
37
|
Vaccination with Live Attenuated Simian Immunodeficiency Virus (SIV) Protects from Mucosal, but Not Necessarily Intravenous, Challenge with a Minimally Heterologous SIV. J Virol 2016; 90:5541-5548. [PMID: 26962218 DOI: 10.1128/jvi.00192-16] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 02/26/2016] [Indexed: 12/14/2022] Open
Abstract
UNLABELLED Few studies have evaluated the impact of the viral challenge route on protection against a heterologous simian immunodeficiency virus (SIV) challenge. We vaccinated seven macaques with a live attenuated SIV that differed from SIVmac239Δnef by 24 amino acids, called m3KOΔnef. All animals were protected from an intrarectal SIVmac239 challenge, whereas only four animals were protected from subsequent intravenous SIVmac239 challenge. These data suggest that immune responses elicited by vaccination with live attenuated SIV in an individual animal can confer protection from intrarectal challenge while remaining insufficient for protection from intravenous challenge. IMPORTANCE Our study is important because we show that vaccinated animals can be protected from a mucosal challenge with a heterologous SIV, but the same animals are not necessarily protected from intravenous challenge with the same virus. This is unique because in most studies, either vaccinated animals are challenged multiple times by the same route or only a single challenge is performed. An individually vaccinated animal is rarely challenged multiple times by different routes, so protection from different challenge routes cannot be measured in the same animal. Our data imply that vaccine-elicited responses in an individual animal may be insufficient for protection from intravenous challenge but may be suitable for protection from a mucosal challenge that better approximates human immunodeficiency virus (HIV) exposure.
Collapse
|
38
|
Affiliation(s)
- Akiko Iwasaki
- Howard Hughes Medical Institute, Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520;
| |
Collapse
|
39
|
Archary D, Seaton KE, Passmore JS, Werner L, Deal A, Dunphy LJ, Arnold KB, Yates NL, Lauffenburger DA, Bergin P, Liebenberg LJ, Samsunder N, Mureithi MW, Altfeld M, Garrett N, Karim QA, Karim SSA, Morris L, Tomaras GD. Distinct genital tract HIV-specific antibody profiles associated with tenofovir gel. Mucosal Immunol 2016; 9:821-833. [PMID: 26813340 PMCID: PMC4848129 DOI: 10.1038/mi.2015.145] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 11/30/2015] [Indexed: 02/04/2023]
Abstract
The impact of topical antiretrovirals for pre-exposure prophylaxis on humoral responses following HIV infection is unknown. Using a binding antibody multiplex assay, we investigated HIV-specific IgG and IgA responses to envelope glycoproteins, p24 Gag and p66, in the genital tract (GT) and plasma following HIV acquisition in women assigned to tenofovir gel (n=24) and placebo gel (n=24) in the CAPRISA 004 microbicide trial to assess if this topical antiretroviral had an impact on mucosal and systemic antibody responses. Linear mixed effect modeling and partial least squares discriminant analysis was used to identify multivariate antibody signatures associated with tenofovir use. There were significantly higher response rates to gp120 Env (P=0.03), p24 (P=0.002), and p66 (P=0.009) in plasma and GT in women assigned to tenofovir than placebo gel at multiple time points post infection. Notably, p66 IgA titers in the GT and plasma were significantly higher in the tenofovir compared with the placebo arm (P<0.05). Plasma titers for 9 of the 10 HIV-IgG specificities predicted GT levels. Taken together, these data suggest that humoral immune responses are increased in blood and GT of individuals who acquire HIV infection in the presence of tenofovir gel.
Collapse
Affiliation(s)
- D Archary
- Centre for the AIDS Program of Research in South Africa, University of KwaZulu-Natal, Durban, South Africa
| | - KE Seaton
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - JS Passmore
- Centre for the AIDS Program of Research in South Africa, University of KwaZulu-Natal, Durban, South Africa
- Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - L Werner
- Centre for the AIDS Program of Research in South Africa, University of KwaZulu-Natal, Durban, South Africa
| | - A Deal
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - LJ Dunphy
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - KB Arnold
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - NL Yates
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - DA Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - P Bergin
- Imperial College, International AIDS Vaccine Initiative Core Immune Monitoring Laboratory, London, UK
| | - LJ Liebenberg
- Centre for the AIDS Program of Research in South Africa, University of KwaZulu-Natal, Durban, South Africa
| | - N Samsunder
- Centre for the AIDS Program of Research in South Africa, University of KwaZulu-Natal, Durban, South Africa
| | - MW Mureithi
- KAVI Institute of Clinical Research, School of Medicine, College of Health Sciences, University of Nairobi, Nairobi, Kenya
| | - M Altfeld
- Heinrich-Pette Institut, Leibniz Institute for Experimental Virology, University of Hamburg, Hamburg, Germany
| | - N Garrett
- Centre for the AIDS Program of Research in South Africa, University of KwaZulu-Natal, Durban, South Africa
| | - Q Abdool Karim
- Centre for the AIDS Program of Research in South Africa, University of KwaZulu-Natal, Durban, South Africa
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - SS Abdool Karim
- Centre for the AIDS Program of Research in South Africa, University of KwaZulu-Natal, Durban, South Africa
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - L Morris
- Centre for the AIDS Program of Research in South Africa, University of KwaZulu-Natal, Durban, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - GD Tomaras
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
- Departments of Surgery, Immunology and Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| |
Collapse
|
40
|
Mkhize NN, Durgiah R, Ashley V, Archary D, Garrett NJ, Karim QA, Karim SSA, Moore PL, Yates N, Passmore JAS, Tomaras GD, Morris L. Broadly neutralizing antibody specificities detected in the genital tract of HIV-1 infected women. AIDS 2016; 30:1005-14. [PMID: 26836790 PMCID: PMC4816677 DOI: 10.1097/qad.0000000000001038] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Broadly neutralizing antibodies (bNAbs) targeting conserved epitopes on the HIV envelope glycoprotein have been identified in blood from HIV-1 infected women. We investigated whether antibodies in the genital tract from these women share similar epitope specificities and functional profiles as those in blood. METHODS Immunoglobulin (Ig)G and IgA antibodies were isolated from cervicovaginal lavages or Softcups from 13 HIV-infected women in the CAPRISA cohort using Protein G and Peptide M, respectively. Binding antibodies to envelope antigens were quantified by ELISA and binding antibody multiplex assay. Neutralizing antibody titers and epitope targets were measured using the TZM-bl assay with Env-pseudotyped wild-type and mutated viruses. RESULTS HIV-specific IgG, but not IgA, was detected in genital secretions and the ratio of total IgG to HIV-specific IgG was similar to plasma. HIV-specific IgG reacted with multiple envelope antigens, including V1V2, gp120, gp140 and gp41. Two women had high plasma titers of HIV-specific IgG3 which was also detected in their genital tract samples. IgG from the genital tract had neutralizing activity against both Tier 1 and Tier 2 primary HIV-isolates. Antibodies targeting well known glycan epitopes and the membrane proximal region of gp41 were detected in genital secretions, and matched specificities in plasma. CONCLUSIONS Women with plasma bNAbs have overlapping specificities in their genital secretions, indicating that these predominantly IgG isotype antibodies may transudate from blood to the genital tract. These data provide evidence that induction of systemic HIV-specific bNAbs can lead to antiviral immunity at the portal of entry.
Collapse
Affiliation(s)
- Nonhlanhla N. Mkhize
- National Institute for Communicable Diseases, Johannesburg, South Africa
- University of the Witwatersrand, Johannesburg, South Africa
| | - Raveshni Durgiah
- National Institute for Communicable Diseases, Johannesburg, South Africa
| | - Vicki Ashley
- Duke Human Vaccine Institute and Departments of Surgery, Immunology and Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina 27710
| | - Derseree Archary
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
| | - Nigel J. Garrett
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
| | - Quarraisha Abdool Karim
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
| | - Salim S. Abdool Karim
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
| | - Penny L. Moore
- National Institute for Communicable Diseases, Johannesburg, South Africa
- University of the Witwatersrand, Johannesburg, South Africa
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
| | - Nicole Yates
- Duke Human Vaccine Institute and Departments of Surgery, Immunology and Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina 27710
| | - Jo-Ann S. Passmore
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
- University of Cape Town, Cape Town, South Africa
- National Health Laboratory Services, South Africa
| | - Georgia D. Tomaras
- Duke Human Vaccine Institute and Departments of Surgery, Immunology and Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina 27710
| | - Lynn Morris
- National Institute for Communicable Diseases, Johannesburg, South Africa
- University of the Witwatersrand, Johannesburg, South Africa
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
41
|
Hodara VL, Parodi LM, Keckler MS, Giavedoni LD. Increases in NKG2C Expression on T Cells and Higher Levels of Circulating CD8 + B Cells Are Associated with Sterilizing Immunity Provided by a Live Attenuated SIV Vaccine. AIDS Res Hum Retroviruses 2016; 32:1125-1134. [PMID: 26986800 DOI: 10.1089/aid.2015.0300] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Vaccines based on live attenuated viruses are highly effective immunogens in the simian immunodeficiency virus (SIV)/rhesus macaque animal model and offer the possibility of studying correlates of protection against infection with virulent virus. We utilized a tether system for studying, in naive macaques and animals vaccinated with a live-attenuated vaccine, the acute events after challenge with pathogenic SIV. This approach allowed for the frequent sampling of small blood volumes without sedation or restraining of the animals, thus reducing the confounding effect of sampling stress. Before challenge, vaccinated animals presented significantly higher levels of proliferating and activated B cells than naive macaques, which were manifested by high expression of CD8 on B cells. After SIV challenge, the only changes observed in protected vaccinated macaques were significant increases in expression of the NK marker NKG2C on CD4 and CD8 T cells. We also identified that infection of naive macaques with SIV resulted in a transient peak of expression of CD20 on CD8 T cells and a constant rise in the number of B cells expressing CD8. Finally, analysis of a larger cohort of vaccinated animals identified that, even when circulating levels of vaccine virus are below the limit of detection, live attenuated vaccines induce systemic increases of IP-10 and perforin. These studies indicate that components of both the innate and adaptive immune systems of animals inoculated with a live-attenuated SIV vaccine respond to and control infection with virulent virus. Persistence of the vaccine virus in tissues may explain the elevated cytokine and B-cell activation levels. In addition, our report underpins the utility of the tether system for the intensive study of acute immune responses to viral infections.
Collapse
Affiliation(s)
- Vida L. Hodara
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, Texas
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas
| | - Laura M. Parodi
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, Texas
| | - M. Shannon Keckler
- Division of Healthcare Quality Promotion, Centers for Diseases Control and Prevention, Atlanta, Georgia
| | - Luis D. Giavedoni
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, Texas
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas
| |
Collapse
|
42
|
Zeng M, Smith AJ, Shang L, Wietgrefe SW, Voss JE, Carlis JV, Li Q, Piatak M, Lifson JD, Johnson RP, Haase AT. Mucosal Humoral Immune Response to SIVmac239∆nef Vaccination and Vaginal Challenge. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 196:2809-18. [PMID: 26864031 PMCID: PMC4779663 DOI: 10.4049/jimmunol.1500156] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 12/31/2015] [Indexed: 01/17/2023]
Abstract
Live attenuated vaccines such as SIV with a deleted nef gene have provided the most robust protection against subsequent vaginal challenge with wild-type (WT) SIV in the SIV-rhesus macaque model of HIV-1 transmission to women. Hence, identifying correlates of this protection could enable design of an effective HIV-1 vaccine. One such prechallenge correlate of protection from vaginal challenge has recently been identified as a system with three components: 1) IgG Abs reacting with the viral envelope glycoprotein trimeric gp41; 2) produced by plasma cells in the submucosa and ectopic tertiary lymphoid follicles in the ectocervix and vagina; and 3) concentrated on the path of virus entry by the neonatal FcR in the overlying epithelium. We now examine the mucosal production of the Ab component of this system after vaginal challenge. We show that vaginal challenge immediately elicits striking increases in plasma cells not only in the female reproductive tract but also at other mucosal sites, and that these increases correlate with low but persistent replication at mucosal sites. We describe vaginal ectopic follicles that are structurally and functionally organized similar to follicles in secondary lymphoid organs, and we provide inferential evidence for a key role of the female reproductive tract epithelium in facilitating Ab production, affinity maturation, and class switch recombination. Vaccination thus accesses an epithelial-immune system axis in the female reproductive tract to respond to exposure to mucosal pathogens. Designing strategies to mimic this system could advance development of an effective HIV-1 vaccine.
Collapse
Affiliation(s)
- Ming Zeng
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, MN 55455
| | - Anthony J Smith
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, MN 55455
| | - Liang Shang
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, MN 55455
| | - Stephen W Wietgrefe
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, MN 55455
| | - James E Voss
- Department of Immunology and Microbial Science, International AIDS Vaccine Initiative Neutralizing Antibody Center, Center for HIV/AIDS Vaccine Immunology and Immunogen Design, The Scripps Research Institute, La Jolla, CA 92037
| | - John V Carlis
- Department of Computer Science and Engineering, College of Science and Engineering, University of Minnesota, Minneapolis, MN 55455
| | - Qingsheng Li
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, MN 55455
| | - Michael Piatak
- AIDS and Cancer Virus Program, Science Applications International Corporation-Frederick, Inc., National Cancer Institute, Frederick, MD 21702
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Science Applications International Corporation-Frederick, Inc., National Cancer Institute, Frederick, MD 21702
| | - R Paul Johnson
- New England Primate Research Center, Harvard Medical School, Southborough, MA, 01772; and Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139
| | - Ashley T Haase
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, MN 55455;
| |
Collapse
|
43
|
Muller S, Parsons MS, Kohler H, Grant M. The Significance of a Common Idiotype (1F7) on Antibodies against Human Immune Deficiency Virus Type 1 and Hepatitis C Virus. Front Oncol 2016; 6:11. [PMID: 26904499 PMCID: PMC4742788 DOI: 10.3389/fonc.2016.00011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 01/12/2016] [Indexed: 11/13/2022] Open
Abstract
In this review, we trace the concept and potential functional role of regulatory idiotypes in the immune response to human immunodeficiency virus type 1 (HIV-1), simian immunodeficiency virus, and hepatitis C virus (HCV). A major idiotype involved in these viral infections is recognized and defined by a murine monoclonal antibody (1F7). Antibodies expressing the idiotype defined by 1F7 are dominant in HIV-1 infection and are also found on many broadly neutralizing antibodies against HIV-1. This regulatory idiotypic axis offers opportunities for exploitation in vaccine development for HIV-1, HCV, and other chronic viral infections.
Collapse
Affiliation(s)
| | - Matthew S Parsons
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne , Melbourne, VIC , Australia
| | - Heinz Kohler
- Department of Microbiology and Immunology, University of Kentucky , Lexington, KY , USA
| | - Michael Grant
- Immunology and Infectious Diseases Program, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland , St. John's, NL , Canada
| |
Collapse
|
44
|
Prospects for engineering HIV-specific antibodies for enhanced effector function and half-life. Curr Opin HIV AIDS 2016; 10:160-9. [PMID: 25700208 DOI: 10.1097/coh.0000000000000149] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
PURPOSE OF REVIEW A wealth of recent animal model data suggests that as exciting possibilities for the use of antibodies in passive immunotherapy strategies continue to develop, it will be important to broadly consider how antibodies achieve anti-HIV-1 effect in vivo. RECENT FINDINGS Beyond neutralization breadth and potency, substantial evidence from natural infection, vaccination, and studies in animal models points to a critical role for antibody Fc receptor (FcR) engagement in reducing risk of infection, decreasing postinfection viremia, and delaying viral rebound. Supporting these findings in the setting of HIV, the clinical maturation of recombinant antibody therapeutics has reinforced the importance of Fc-driven activity in vivo across many disease settings, as well as opportunely resulted in the development and exploration of a number of engineered Fc sequence and glycosylation variants that possess differential binding to FcRs. Exploiting these variants as tools, the individual and concerted effects of antibody effector functions such as antibody-dependent cellular cytotoxicity, antibody-dependent cell-mediated virus inhibition, phagocytosis, complement-dependent cytotoxicity, antibody half-life, and compartmentalization are now being explored. As exciting molecular therapies are advanced, these studies promise to provide insight into optimal in-vivo antibody activity profiles. SUMMARY Careful consideration of recent progress in understanding protective antibody activities in vivo can point toward how tailoring antibody activity via Fc domain modification may enable optimization of HIV prevention and eradication strategies.
Collapse
|
45
|
Lelièvre JD, Lévy Y. HIV-1 prophylactic vaccines: state of the art. J Virus Erad 2016; 2:5-11. [PMID: 27482428 PMCID: PMC4946697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The quest for an effective HIV-1 vaccine began early in the course of the HIV pandemic. Over time, the paradigm has evolved from B cell- towards T cell-based vaccines. Results from initial Phase II/III trials have been disappointing; however, while modest, the unexpected results of the Phase II/III RV144 trial in Thailand have re-energised the field. Indeed a clear correlation was demonstrated in this trial between protection and immunological biomarkers, namely non-neutralising antibodies against the V1V2 region. Recent data obtained from cohorts of recently HIV-1-infected individuals have enabled exploration of the role of neutralising antibodies and their potential use in HIV-1 prevention. Results from non-human primate models using a cytomegalovirus vector have also shown the potential for a prophylactic HIV vaccine to induce effective T cell responses. Finally, the development of new vaccine vectors and trial strategies has also allowed progress in the field. Therefore, HIV-1 vaccine research remains a dynamic field that has also been stimulated by the recent positive results of pre-exposure prophylaxis strategies with antiretrovirals.
Collapse
Affiliation(s)
- Jean-Daniel Lelièvre
- AP-HP, Hôpital Henri Mondor – Albert Chenevier, Service d’Immunologie Clinique et Maladies Infectieuses, Créteil, 94000, France,Corresponding author: Jean-Daniel Lelièvre, Service d’Immunologie Clinique et Maladies Infectieuses, CHU Henri Mondor, 51 avenue Mal de Lattre de Tassigny, 94010, Créteil, France
| | - Yves Lévy
- AP-HP, Hôpital Henri Mondor – Albert Chenevier, Service d’Immunologie Clinique et Maladies Infectieuses, Créteil, 94000, France
| |
Collapse
|
46
|
|
47
|
Santra S, Tomaras GD, Warrier R, Nicely NI, Liao HX, Pollara J, Liu P, Alam SM, Zhang R, Cocklin SL, Shen X, Duffy R, Xia SM, Schutte RJ, Pemble IV CW, Dennison SM, Li H, Chao A, Vidnovic K, Evans A, Klein K, Kumar A, Robinson J, Landucci G, Forthal DN, Montefiori DC, Kaewkungwal J, Nitayaphan S, Pitisuttithum P, Rerks-Ngarm S, Robb ML, Michael NL, Kim JH, Soderberg KA, Giorgi EE, Blair L, Korber BT, Moog C, Shattock RJ, Letvin NL, Schmitz JE, Moody MA, Gao F, Ferrari G, Shaw GM, Haynes BF. Human Non-neutralizing HIV-1 Envelope Monoclonal Antibodies Limit the Number of Founder Viruses during SHIV Mucosal Infection in Rhesus Macaques. PLoS Pathog 2015; 11:e1005042. [PMID: 26237403 PMCID: PMC4523205 DOI: 10.1371/journal.ppat.1005042] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Accepted: 06/23/2015] [Indexed: 11/19/2022] Open
Abstract
HIV-1 mucosal transmission begins with virus or virus-infected cells moving through mucus across mucosal epithelium to infect CD4+ T cells. Although broadly neutralizing antibodies (bnAbs) are the type of HIV-1 antibodies that are most likely protective, they are not induced with current vaccine candidates. In contrast, antibodies that do not neutralize primary HIV-1 strains in the TZM-bl infection assay are readily induced by current vaccine candidates and have also been implicated as secondary correlates of decreased HIV-1 risk in the RV144 vaccine efficacy trial. Here, we have studied the capacity of anti-Env monoclonal antibodies (mAbs) against either the immunodominant region of gp41 (7B2 IgG1), the first constant region of gp120 (A32 IgG1), or the third variable loop (V3) of gp120 (CH22 IgG1) to modulate in vivo rectal mucosal transmission of a high-dose simian-human immunodeficiency virus (SHIV-BaL) in rhesus macaques. 7B2 IgG1 or A32 IgG1, each containing mutations to enhance Fc function, was administered passively to rhesus macaques but afforded no protection against productive clinical infection while the positive control antibody CH22 IgG1 prevented infection in 4 of 6 animals. Enumeration of transmitted/founder (T/F) viruses revealed that passive infusion of each of the three antibodies significantly reduced the number of T/F genomes. Thus, some antibodies that bind HIV-1 Env but fail to neutralize virus in traditional neutralization assays may limit the number of T/F viruses involved in transmission without leading to enhancement of viral infection. For one of these mAbs, gp41 mAb 7B2, we provide the first co-crystal structure in complex with a common cyclical loop motif demonstrated to be critical for infection by other retroviruses.
Collapse
Affiliation(s)
- Sampa Santra
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail: (SS); (GDT); (BFH)
| | - Georgia D. Tomaras
- Duke Human Vaccine Institute, Duke School of Medicine, Durham, North Carolina, United States of America
- * E-mail: (SS); (GDT); (BFH)
| | - Ranjit Warrier
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Nathan I. Nicely
- Duke Human Vaccine Institute, Duke School of Medicine, Durham, North Carolina, United States of America
| | - Hua-Xin Liao
- Duke Human Vaccine Institute, Duke School of Medicine, Durham, North Carolina, United States of America
| | - Justin Pollara
- Duke Human Vaccine Institute, Duke School of Medicine, Durham, North Carolina, United States of America
| | - Pinghuang Liu
- Duke Human Vaccine Institute, Duke School of Medicine, Durham, North Carolina, United States of America
| | - S. Munir Alam
- Duke Human Vaccine Institute, Duke School of Medicine, Durham, North Carolina, United States of America
| | - Ruijun Zhang
- Duke Human Vaccine Institute, Duke School of Medicine, Durham, North Carolina, United States of America
| | - Sarah L. Cocklin
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Xiaoying Shen
- Duke Human Vaccine Institute, Duke School of Medicine, Durham, North Carolina, United States of America
| | - Ryan Duffy
- Duke Human Vaccine Institute, Duke School of Medicine, Durham, North Carolina, United States of America
| | - Shi-Mao Xia
- Duke Human Vaccine Institute, Duke School of Medicine, Durham, North Carolina, United States of America
| | - Robert J. Schutte
- Duke Human Vaccine Institute, Duke School of Medicine, Durham, North Carolina, United States of America
| | - Charles W. Pemble IV
- Duke Human Vaccine Institute, Duke School of Medicine, Durham, North Carolina, United States of America
| | - S. Moses Dennison
- Duke Human Vaccine Institute, Duke School of Medicine, Durham, North Carolina, United States of America
| | - Hui Li
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Andrew Chao
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Kora Vidnovic
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Abbey Evans
- Department of Medicine, St Mary’s Campus, Imperial College London, London, United Kingdom
| | - Katja Klein
- Department of Medicine, St Mary’s Campus, Imperial College London, London, United Kingdom
| | - Amit Kumar
- Duke Human Vaccine Institute, Duke School of Medicine, Durham, North Carolina, United States of America
| | - James Robinson
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Gary Landucci
- Division of Infectious Diseases, Department of Medicine, University of California, Irvine, Irvine, California, United States of America
| | - Donald N. Forthal
- Division of Infectious Diseases, Department of Medicine, University of California, Irvine, Irvine, California, United States of America
| | - David C. Montefiori
- Duke Human Vaccine Institute, Duke School of Medicine, Durham, North Carolina, United States of America
| | | | - Sorachai Nitayaphan
- Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand
| | | | | | - Merlin L. Robb
- US Military Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Nelson L. Michael
- US Military Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Jerome H. Kim
- US Military Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Kelly A. Soderberg
- Duke Human Vaccine Institute, Duke School of Medicine, Durham, North Carolina, United States of America
| | - Elena E. Giorgi
- Theoretical Division, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Lily Blair
- Theoretical Division, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Bette T. Korber
- Theoretical Division, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Christiane Moog
- U1109, INSERM University of Strasbourg, Strasbourg, Alsace, France
| | - Robin J. Shattock
- Department of Medicine, St Mary’s Campus, Imperial College London, London, United Kingdom
| | - Norman L. Letvin
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Joern E. Schmitz
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - M. A. Moody
- Duke Human Vaccine Institute, Duke School of Medicine, Durham, North Carolina, United States of America
| | - Feng Gao
- Duke Human Vaccine Institute, Duke School of Medicine, Durham, North Carolina, United States of America
| | - Guido Ferrari
- Duke Human Vaccine Institute, Duke School of Medicine, Durham, North Carolina, United States of America
| | - George M. Shaw
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Barton F. Haynes
- Duke Human Vaccine Institute, Duke School of Medicine, Durham, North Carolina, United States of America
- * E-mail: (SS); (GDT); (BFH)
| |
Collapse
|
48
|
Williams WB, Liao HX, Moody MA, Kepler TB, Alam SM, Gao F, Wiehe K, Trama AM, Jones K, Zhang R, Song H, Marshall DJ, Whitesides JF, Sawatzki K, Hua A, Liu P, Tay MZ, Seaton KE, Shen X, Foulger A, Lloyd KE, Parks R, Pollara J, Ferrari G, Yu JS, Vandergrift N, Montefiori DC, Sobieszczyk ME, Hammer S, Karuna S, Gilbert P, Grove D, Grunenberg N, McElrath MJ, Mascola JR, Koup RA, Corey L, Nabel GJ, Morgan C, Churchyard G, Maenza J, Keefer M, Graham BS, Baden LR, Tomaras GD, Haynes BF. HIV-1 VACCINES. Diversion of HIV-1 vaccine-induced immunity by gp41-microbiota cross-reactive antibodies. Science 2015; 349:aab1253. [PMID: 26229114 DOI: 10.1126/science.aab1253] [Citation(s) in RCA: 161] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 07/09/2015] [Indexed: 01/04/2023]
Abstract
An HIV-1 DNA prime vaccine, with a recombinant adenovirus type 5 (rAd5) boost, failed to protect from HIV-1 acquisition. We studied the nature of the vaccine-induced antibody (Ab) response to HIV-1 envelope (Env). HIV-1-reactive plasma Ab titers were higher to Env gp41 than to gp120, and repertoire analysis demonstrated that 93% of HIV-1-reactive Abs from memory B cells responded to Env gp41. Vaccine-induced gp41-reactive monoclonal antibodies were non-neutralizing and frequently polyreactive with host and environmental antigens, including intestinal microbiota (IM). Next-generation sequencing of an immunoglobulin heavy chain variable region repertoire before vaccination revealed an Env-IM cross-reactive Ab that was clonally related to a subsequent vaccine-induced gp41-reactive Ab. Thus, HIV-1 Env DNA-rAd5 vaccine induced a dominant IM-polyreactive, non-neutralizing gp41-reactive Ab repertoire response that was associated with no vaccine efficacy.
Collapse
Affiliation(s)
- Wilton B Williams
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA.
| | - Hua-Xin Liao
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - M Anthony Moody
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Thomas B Kepler
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA
| | - S Munir Alam
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Feng Gao
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Kevin Wiehe
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Ashley M Trama
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Kathryn Jones
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Ruijun Zhang
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Hongshuo Song
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Dawn J Marshall
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - John F Whitesides
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Kaitlin Sawatzki
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA
| | - Axin Hua
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA
| | - Pinghuang Liu
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Matthew Z Tay
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Kelly E Seaton
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Xiaoying Shen
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Andrew Foulger
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Krissey E Lloyd
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Robert Parks
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Justin Pollara
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Guido Ferrari
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Jae-Sung Yu
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Nathan Vandergrift
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - David C Montefiori
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | | | - Scott Hammer
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Shelly Karuna
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Peter Gilbert
- The Statistical Center for HIV/AIDS Research and Prevention (SCHARP), Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Doug Grove
- The Statistical Center for HIV/AIDS Research and Prevention (SCHARP), Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Nicole Grunenberg
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - M Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Richard A Koup
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Gary J Nabel
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Cecilia Morgan
- The Statistical Center for HIV/AIDS Research and Prevention (SCHARP), Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Janine Maenza
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Michael Keefer
- University of Rochester School of Medicine, Rochester, NY, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Georgia D Tomaras
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Barton F Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
49
|
Kohler H. Novel vaccine concept based on back-boost effect in viral infection. Vaccine 2015; 33:3274-5. [PMID: 26032350 DOI: 10.1016/j.vaccine.2015.05.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 05/14/2015] [Accepted: 05/18/2015] [Indexed: 11/30/2022]
Abstract
A novel vaccine concept is discussed based on recent evidence of a "back-boost" effect in Influenza infection. The initial immune response to the infection is imprinted through an immune memory pathway. The immune memory in the back-boost mechanism could be used in reversed order as a "forward-boost" in the proposed vaccine concept.
Collapse
Affiliation(s)
- Heinz Kohler
- Univ. of Kentucky, Microbiology/Immunology, 800 Rose Street, Lexington, KY 40536, United States.
| |
Collapse
|
50
|
Dispelling myths and focusing on notable concepts in HIV pathogenesis. Trends Mol Med 2015; 21:341-53. [PMID: 25883070 DOI: 10.1016/j.molmed.2015.03.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 03/05/2015] [Accepted: 03/13/2015] [Indexed: 01/01/2023]
Abstract
Since the discovery of HIV over three decades ago, major efforts have been made to control and perhaps eliminate HIV infection worldwide. During these studies, certain myths or misconceptions about this infectious disease have been emphasized and other potentially beneficial concepts have received less attention. A true long-term solution to HIV infection merits an appreciation of alternative ideas and findings that could be beneficial in the ultimate control of HIV/AIDS. Here, I discuss six issues and call for more attention to the science of HIV and well-designed clinical trials.
Collapse
|