1
|
Berkson JD, Wate CE, Allen GB, Schubert AM, Dunbar KE, Coryell MP, Sava RL, Gao Y, Hastie JL, Smith EM, Kenneally CR, Zimmermann SK, Carlson PE. Phage-specific immunity impairs efficacy of bacteriophage targeting Vancomycin Resistant Enterococcus in a murine model. Nat Commun 2024; 15:2993. [PMID: 38582763 PMCID: PMC10998888 DOI: 10.1038/s41467-024-47192-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/22/2024] [Indexed: 04/08/2024] Open
Abstract
Bacteriophage therapy is a promising approach to address antimicrobial infections though questions remain regarding the impact of the immune response on clinical effectiveness. Here, we develop a mouse model to assess phage treatment using a cocktail of five phages from the Myoviridae and Siphoviridae families that target Vancomycin-Resistant Enterococcus gut colonization. Phage treatment significantly reduces fecal bacterial loads of Vancomycin-Resistant Enterococcus. We also characterize immune responses elicited following administration of the phage cocktail. While minimal innate responses are observed after phage administration, two rounds of treatment induces phage-specific neutralizing antibodies and accelerate phage clearance from tissues. Interestingly, the myophages in our cocktail induce a more robust neutralizing antibody response than the siphophages. This anti-phage immunity reduces the effectiveness of the phage cocktail in our murine model. Collectively, this study shows phage-specific immune responses may be an important consideration in the development of phage cocktails for therapeutic use.
Collapse
Affiliation(s)
- Julia D Berkson
- Food and Drug Administration, Center for Biologics Evaluation and Research, Office of Vaccines Research and Review, Division of Bacterial Parasitic and Allergenic Products, Laboratory of Mucosal Pathogens and Cellular Immunology, 10903 New Hampshire Ave, Silver Spring, MD, 20832, USA
| | - Claire E Wate
- Food and Drug Administration, Center for Biologics Evaluation and Research, Office of Vaccines Research and Review, Division of Bacterial Parasitic and Allergenic Products, Laboratory of Mucosal Pathogens and Cellular Immunology, 10903 New Hampshire Ave, Silver Spring, MD, 20832, USA
| | - Garrison B Allen
- Food and Drug Administration, Center for Biologics Evaluation and Research, Office of Vaccines Research and Review, Division of Bacterial Parasitic and Allergenic Products, Laboratory of Mucosal Pathogens and Cellular Immunology, 10903 New Hampshire Ave, Silver Spring, MD, 20832, USA
| | - Alyxandria M Schubert
- Food and Drug Administration, Center for Biologics Evaluation and Research, Office of Vaccines Research and Review, Division of Bacterial Parasitic and Allergenic Products, Laboratory of Mucosal Pathogens and Cellular Immunology, 10903 New Hampshire Ave, Silver Spring, MD, 20832, USA
| | - Kristin E Dunbar
- Food and Drug Administration, Center for Biologics Evaluation and Research, Office of Vaccines Research and Review, Division of Bacterial Parasitic and Allergenic Products, Laboratory of Mucosal Pathogens and Cellular Immunology, 10903 New Hampshire Ave, Silver Spring, MD, 20832, USA
| | - Michael P Coryell
- Food and Drug Administration, Center for Biologics Evaluation and Research, Office of Vaccines Research and Review, Division of Bacterial Parasitic and Allergenic Products, Laboratory of Mucosal Pathogens and Cellular Immunology, 10903 New Hampshire Ave, Silver Spring, MD, 20832, USA
| | - Rosa L Sava
- Food and Drug Administration, Center for Biologics Evaluation and Research, Office of Vaccines Research and Review, Division of Bacterial Parasitic and Allergenic Products, Laboratory of Mucosal Pathogens and Cellular Immunology, 10903 New Hampshire Ave, Silver Spring, MD, 20832, USA
| | - Yamei Gao
- Food and Drug Administration, Center for Biologics Evaluation and Research, Office of Vaccines Research and Review, Division of Viral Products, Laboratory of Pediatric and Respiratory Viral Diseases, 10903 New Hampshire Ave, Silver Spring, MD, 20832, USA
| | - Jessica L Hastie
- Food and Drug Administration, Center for Biologics Evaluation and Research, Office of Vaccines Research and Review, Division of Bacterial Parasitic and Allergenic Products, Laboratory of Mucosal Pathogens and Cellular Immunology, 10903 New Hampshire Ave, Silver Spring, MD, 20832, USA
| | - Emily M Smith
- Food and Drug Administration, Center for Biologics Evaluation and Research, Office of Vaccines Research and Review, Division of Bacterial Parasitic and Allergenic Products, Laboratory of Mucosal Pathogens and Cellular Immunology, 10903 New Hampshire Ave, Silver Spring, MD, 20832, USA
| | - Charlotte R Kenneally
- Food and Drug Administration, Center for Biologics Evaluation and Research, Office of Vaccines Research and Review, Division of Bacterial Parasitic and Allergenic Products, Laboratory of Mucosal Pathogens and Cellular Immunology, 10903 New Hampshire Ave, Silver Spring, MD, 20832, USA
| | - Sally K Zimmermann
- Food and Drug Administration, Center for Biologics Evaluation and Research, Office of Vaccines Research and Review, Division of Bacterial Parasitic and Allergenic Products, Laboratory of Mucosal Pathogens and Cellular Immunology, 10903 New Hampshire Ave, Silver Spring, MD, 20832, USA
| | - Paul E Carlson
- Food and Drug Administration, Center for Biologics Evaluation and Research, Office of Vaccines Research and Review, Division of Bacterial Parasitic and Allergenic Products, Laboratory of Mucosal Pathogens and Cellular Immunology, 10903 New Hampshire Ave, Silver Spring, MD, 20832, USA.
| |
Collapse
|
2
|
Li Z, Zhu Y, Yan F, Jin H, Wang Q, Zhao Y, Feng N, Wang T, Li N, Yang S, Xia X, Cong Y. Inactivated Recombinant Rabies Virus Displaying the Nipah Virus Envelope Glycoproteins Induces Systemic Immune Responses in Mice. Vaccines (Basel) 2023; 11:1758. [PMID: 38140162 PMCID: PMC10747385 DOI: 10.3390/vaccines11121758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/07/2023] [Accepted: 11/11/2023] [Indexed: 12/24/2023] Open
Abstract
Nipah virus (NiV) causes severe, lethal encephalitis in humans and pigs. However, there is no licensed vaccine available to prevent NiV infection. In this study, we used the reverse genetic system based on the attenuated rabies virus strain SRV9 to construct two recombinant viruses, rSRV9-NiV-F and rSRV9-NiV-G, which displayed the NiV envelope glycoproteins F and G, respectively. Following three immunizations in BALB/c mice, the inactivated rSRV9-NiV-F and rSRV9-NiV-G alone or in combination, mixed with the adjuvants ISA 201 VG and poly (I:C), were able to induce the antigen-specific cellular and Th1-biased humoral immune responses. The specific antibodies against rSRV9-NiV-F and rSRV9-NiV-G had reactivity with two constructed bacterial-like particles displaying the F and G antigens of NiV. These data demonstrate that rSRV9-NiV-F or rSRV9-NiV-G has the potential to be developed into a promising vaccine candidate against NiV infection.
Collapse
Affiliation(s)
- Zhengrong Li
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun 130122, China
| | - Yanting Zhu
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun 130122, China
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China
| | - Hongli Jin
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun 130122, China
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China
| | - Qi Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China
| | - Yongkun Zhao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China
| | - Na Feng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China
| | - Tiecheng Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China
| | - Nan Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China
| | - Songtao Yang
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun 130122, China
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China
| | - Xianzhu Xia
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun 130122, China
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China
| | - Yanlong Cong
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun 130122, China
| |
Collapse
|
3
|
Henriques P, Rosa A, Caldeira-Araújo H, Soares P, Vigário AM. Flying under the radar - impact and factors influencing asymptomatic DENV infections. Front Cell Infect Microbiol 2023; 13:1284651. [PMID: 38076464 PMCID: PMC10704250 DOI: 10.3389/fcimb.2023.1284651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
The clinical outcome of DENV and other Flaviviruses infections represents a spectrum of severity that ranges from mild manifestations to severe disease, which can ultimately lead to death. Nonetheless, most of these infections result in an asymptomatic outcome that may play an important role in the persistent circulation of these viruses. Also, although little is known about the mechanisms that lead to these asymptomatic infections, they are likely the result of a complex interplay between viral and host factors. Specific characteristics of the infecting viral strain, such as its replicating efficiency, coupled with host factors, like gene expression of key molecules involved in the immune response or in the protection against disease, are among crucial factors to study. This review revisits recent data on factors that may contribute to the asymptomatic outcome of the world's widespread DENV, highlighting the importance of silent infections in the transmission of this pathogen and the immune status of the host.
Collapse
Affiliation(s)
- Paulo Henriques
- Projecto Medicina, Faculdade de Ciências da Vida, Universidade da Madeira, Funchal, Portugal
| | - Alexandra Rosa
- Projecto Medicina, Faculdade de Ciências da Vida, Universidade da Madeira, Funchal, Portugal
| | - Helena Caldeira-Araújo
- Projecto Medicina, Faculdade de Ciências da Vida, Universidade da Madeira, Funchal, Portugal
- CQM-Centro de Química da Madeira, Universidade da Madeira, Funchal, Portugal
| | - Pedro Soares
- Department of Biology, CBMA (Centre of Molecular and Environmental Biology), Braga, Portugal
- Department of Biology, Institute of Science and Innovation for Bio-Sustainability (IB-S), University of Minho, Braga, Portugal
| | - Ana Margarida Vigário
- Projecto Medicina, Faculdade de Ciências da Vida, Universidade da Madeira, Funchal, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
4
|
Arora P, Jain A, Kumar A. Phage design and directed evolution to evolve phage for therapy. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 200:103-126. [PMID: 37739551 DOI: 10.1016/bs.pmbts.2023.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/24/2023]
Abstract
Phage therapy or Phage treatment is the use of bacteriolysing phage in treating bacterial infections by using the viruses that infects and kills bacteria. This technique has been studied and practiced very long ago, but with the advent of antibiotics, it has been neglected. This foregone technique is now witnessing a revival due to development of bacterial resistance. Nowadays, with the awareness of genetic sequence of organisms, it is required that informed choices of phages have to be made for the most efficacious results. Furthermore, phages with the evolving genes are taken into consideration for the subsequent improvement in treating the patients for bacterial diseases. In addition, direct evolution methods are increasingly developing, since these are capable of creating new biological molecules having changed or unique activities, such as, improved target specificity, evolution of novel proteins with new catalytic properties or creation of nucleic acids that are capable of recognizing required pathogenic bacteria. This system is incorporates continuous evolution such as protein or genes are put under continuous evolution by providing continuous mutagenesis with least human intervention. Although, this system providing continuous directed evolution is very effective, it imposes some challenges due to requirement of heavy investment of time and resources. This chapter focuses on development of phage as a therapeutic agent against various bacteria causing diseases and it improvement using direct evolution of proteins and nucleic acids such that they target specific organisms.
Collapse
Affiliation(s)
- Priyancka Arora
- Department of Biotechnology, Faculty of Engineering and Technology, Rama University, Kanpur, Uttar Pradesh, India
| | - Avni Jain
- Department of Biotechnology, Faculty of Engineering and Technology, Rama University, Kanpur, Uttar Pradesh, India
| | - Ajay Kumar
- Department of Biotechnology, Faculty of Engineering and Technology, Rama University, Kanpur, Uttar Pradesh, India.
| |
Collapse
|
5
|
Repetitive Exposure to Bacteriophage Cocktails against Pseudomonas aeruginosa or Escherichia coli Provokes Marginal Humoral Immunity in Naïve Mice. Viruses 2023; 15:v15020387. [PMID: 36851601 PMCID: PMC9964535 DOI: 10.3390/v15020387] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/18/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
Phage therapy of ventilator-associated pneumonia (VAP) is of great interest due to the rising incidence of multidrug-resistant bacterial pathogens. However, natural or therapy-induced immunity against therapeutic phages remains a potential concern. In this study, we investigated the innate and adaptive immune responses to two different phage cocktails targeting either Pseudomonas aeruginosa or Escherichia coli-two VAP-associated pathogens-in naïve mice without the confounding effects of a bacterial infection. Active or UV-inactivated phage cocktails or buffers were injected intraperitoneally daily for 7 days in C57BL/6J wild-type mice. Blood cell analysis, flow cytometry analysis, assessment of phage distribution and histopathological analysis of spleens were performed at 6 h, 10 days and 21 days after treatment start. Phages reached the lungs and although the phage cocktails were slightly immunogenic, phage injections were well tolerated without obvious adverse effects. No signs of activation of innate or adaptive immune cells were observed; however, both active phage cocktails elicited a minimal humoral response with secretion of phage-specific antibodies. Our findings show that even repetitive injections lead only to a minimal innate and adaptive immune response in naïve mice and suggest that systemic phage treatment is thus potentially suitable for treating bacterial lung infections.
Collapse
|
6
|
Bartsch YC, St Denis KJ, Kaplonek P, Kang J, Lam EC, Burns MD, Farkas EJ, Davis JP, Boribong BP, Edlow AG, Fasano A, Shreffler WG, Zavadska D, Johnson M, Goldblatt D, Balazs AB, Yonker LM, Alter G. SARS-CoV-2 mRNA vaccination elicits robust antibody responses in children. Sci Transl Med 2022; 14:eabn9237. [PMID: 35881018 PMCID: PMC9348753 DOI: 10.1126/scitranslmed.abn9237] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 07/01/2022] [Indexed: 01/11/2023]
Abstract
Although children have been largely spared from coronavirus disease 2019 (COVID-19), the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern (VOCs) with increased transmissibility, combined with fluctuating mask mandates and school reopenings, has led to increased infections and disease among children. Thus, there is an urgent need to roll out COVID-19 vaccines to children of all ages. However, whether children respond equivalently to adults to mRNA vaccines and whether dosing will elicit optimal immunity remain unclear. Here, we aimed to deeply profile the vaccine-induced humoral immune response in 6- to 11-year-old children receiving either a pediatric (50 μg) or adult (100 μg) dose of the mRNA-1273 vaccine and to compare these responses to vaccinated adults, infected children, and children who experienced multisystem inflammatory syndrome in children (MIS-C). Children elicited an IgG-dominant vaccine-induced immune response, surpassing adults at a matched 100-μg dose but more variable immunity at a 50-μg dose. Irrespective of titer, children generated antibodies with enhanced Fc receptor binding capacity. Moreover, like adults, children generated cross-VOC humoral immunity, marked by a decline of omicron-specific receptor binding domain, but robustly preserved omicron spike protein binding. Fc receptor binding capabilities were also preserved in a dose-dependent manner. These data indicate that both the 50- and 100-μg doses of mRNA vaccination in children elicit robust cross-VOC antibody responses and that 100-μg doses in children result in highly preserved omicron-specific functional humoral immunity.
Collapse
Affiliation(s)
- Yannic C Bartsch
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Kerri J St Denis
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Paulina Kaplonek
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Jaewon Kang
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Evan C Lam
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Madeleine D Burns
- Massachusetts General Hospital Department of Pediatrics, Mucosal Immunology and Biology Research Center, Boston, MA 02114, USA
| | - Eva J Farkas
- Massachusetts General Hospital Department of Pediatrics, Mucosal Immunology and Biology Research Center, Boston, MA 02114, USA
| | - Jameson P Davis
- Massachusetts General Hospital Department of Pediatrics, Mucosal Immunology and Biology Research Center, Boston, MA 02114, USA
| | - Brittany P Boribong
- Massachusetts General Hospital Department of Pediatrics, Mucosal Immunology and Biology Research Center, Boston, MA 02114, USA
| | - Andrea G Edlow
- Massachusetts General Hospital Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Vincent Center for Reproductive Biology, Boston, MA 02114, USA
| | - Alessio Fasano
- Massachusetts General Hospital Department of Pediatrics, Mucosal Immunology and Biology Research Center, Boston, MA 02114, USA
| | - Wayne G Shreffler
- Massachusetts General Hospital Food Allergy Center, Division of Pediatric Allergy and Immunology, Boston, MA 02114, USA
| | - Dace Zavadska
- Children's Clinical University Hospital, Riga, LV-1004, Latvia
| | - Marina Johnson
- Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London WC1N 1EH, UK
| | - David Goldblatt
- Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London WC1N 1EH, UK
| | | | - Lael M Yonker
- Massachusetts General Hospital Department of Pediatrics, Mucosal Immunology and Biology Research Center, Boston, MA 02114, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| |
Collapse
|
7
|
IgG targeting distinct seasonal coronavirus- conserved SARS-CoV-2 spike subdomains correlates with differential COVID-19 disease outcomes. Cell Rep 2022; 39:110904. [PMID: 35617962 PMCID: PMC9108089 DOI: 10.1016/j.celrep.2022.110904] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 03/25/2022] [Accepted: 05/11/2022] [Indexed: 11/20/2022] Open
Abstract
Despite SARS-CoV-2 being a "novel" virus, early detection of anti-spike IgG in severe COVID-19 patients may be caused by the amplification of humoral memory responses against seasonal coronaviruses. Here, we examine this phenomenon by characterizing anti-spike IgG responses in non-hospitalized convalescent individuals across a spectrum of COVID-19 severity. We observe that disease severity positively correlates with anti-spike IgG levels, IgG cross-reactivity against other betacoronaviruses (β-CoVs), and FcγR activation. Analysis of IgG targeting β-CoV-conserved and non-conserved immunodominant epitopes within the SARS-CoV-2 spike protein revealed epitope-specific relationships: IgG targeting the conserved heptad repeat (HR) 2 region significantly correlates with milder disease, while targeting the conserved S2'FP region correlates with more severe disease. Furthermore, a lower HR2-to-S2'FP IgG-binding ratio correlates with greater disease severity, with ICU-hospitalized COVID-19 patients showing the lowest HR2/S2'FP ratios. These findings suggest that HR2/S2'FP IgG profiles may predict disease severity and offer insight into protective versus deleterious humoral recall responses.
Collapse
|
8
|
Weiss S, Itri V, Pan R, Jiang X, Luo CC, Morris L, Malherbe DC, Barnette P, Alexander J, Kong XP, Haigwood NL, Hessell AJ, Duerr R, Zolla-Pazner S. Differential V2-directed antibody responses in non-human primates infected with SHIVs or immunized with diverse HIV vaccines. Nat Commun 2022; 13:903. [PMID: 35173151 PMCID: PMC8850611 DOI: 10.1038/s41467-022-28450-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 01/25/2022] [Indexed: 11/24/2022] Open
Abstract
V2p and V2i antibodies (Abs) that are specific for epitopes in the V1V2 region of the HIV gp120 envelope (Env) do not effectively neutralize HIV but mediate Fc-dependent anti-viral activities that have been correlated with protection from, or control of HIV, SIV and SHIV infections. Here, we describe a novel molecular toolbox that allows the discrimination of antigenically and functionally distinct polyclonal V2 Ab responses. We identify different patterns of V2 Ab induction by SHIV infection and three separate vaccine regimens that aid in fine-tuning an optimized immunization protocol for inducing V2p and V2i Abs. We observe no, or weak and sporadic V2p and V2i Abs in non-vaccinated SHIV-infected NHPs, but strong V2p and/or V2i Ab responses after immunization with a V2-targeting vaccine protocol. The V2-focused vaccination is superior to both natural infection and to immunization with whole Env constructs for inducing functional V2p- and V2i-specific responses. Strikingly, levels of V2-directed Abs correlate inversely with Abs specific for peptides of V3 and C5. These data demonstrate that a V1V2-targeting vaccine has advantages over the imprecise targeting of SIV/SHIV infections and of whole Env-based immunization regimens for inducing a more focused functional V2p- and V2i-specific Ab response. Here the authors show that an HIV vaccine in non-human primates that focuses antibodies on the V1V2 region of gp120 is superior to infection or immunization with whole envelope vaccines for inducing V1V2 antibodies with anti-viral functions that correlate with protection.
Collapse
Affiliation(s)
- Svenja Weiss
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vincenza Itri
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ruimin Pan
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Xunqing Jiang
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Christina C Luo
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Lynn Morris
- National Institute for Communicable Diseases, National Health Laboratory Service, Sandringham, Johannesburg, South Africa.,MRC Antibody Research Unit, University of the Witwatersrand, Johannesburg and Center for the AIDS Program of Research in South Africa, Johannesburg, South Africa
| | - Delphine C Malherbe
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA.,University of Texas Medical Branch, Department of Pathology, Galveston National Laboratory, Galveston, TX, USA
| | - Philip Barnette
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | - Jeff Alexander
- PaxVax Corporation, Redwood City, CA, USA.,JL Alexander Research and Development Consulting LLC, San Diego, CA, USA
| | - Xiang-Peng Kong
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Nancy L Haigwood
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | - Ann J Hessell
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | - Ralf Duerr
- Department of Microbiology, New York University School of Medicine, New York, NY, USA
| | - Susan Zolla-Pazner
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Department of Microbiology, Icahn School of Medicine, New York, NY, USA.
| |
Collapse
|
9
|
Bartsch YC, St Denis KJ, Kaplonek P, Kang J, Lam EC, Burns MD, Farkas EJ, Davis JP, Boribong BP, Edlow AG, Fasano A, Shreffler W, Zavadska D, Johnson M, Goldblatt D, Balazs AB, Yonker LM, Alter G. Comprehensive antibody profiling of mRNA vaccination in children. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2021.10.07.463592. [PMID: 35018376 PMCID: PMC8750651 DOI: 10.1101/2021.10.07.463592] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
While children have been largely spared from COVID-19 disease, the emergence of viral variants of concern (VOC) with increased transmissibility, combined with fluctuating mask mandates and school re-openings have led to increased infections and disease among children. Thus, there is an urgent need to roll out COVID-19 vaccines to children of all ages. However, whether children respond equivalently to adults to mRNA vaccines and whether dosing will elicit optimal immunity remains unclear. Given the recent announcement of incomplete immunity induced by the pediatric dose of the BNT162b2 vaccine in young children, here we aimed to deeply profile and compare the vaccine-induced humoral immune response in 6-11 year old children receiving the pediatric (50μg) or adult (100μg) dose of the mRNA-1273 vaccine compared to adults and naturally infected children or children that experienced multi inflammatory syndrome in children (MIS-C) for the first time. Children elicited an IgG dominant vaccine induced immune response, surpassing adults at a matched 100μg dose, but more variable immunity at a 50μg dose. Irrespective of titer, children generated antibodies with enhanced Fc-receptor binding capacity. Moreover, like adults, children generated cross-VOC humoral immunity, marked by a decline of omicron receptor binding domain-binding, but robustly preserved omicron Spike-receptor binding, with robustly preserved Fc-receptor binding capabilities, in a dose dependent manner. These data indicate that while both 50μg and 100μg of mRNA vaccination in children elicits robust cross-VOC antibody responses, 100ug of mRNA in children results in highly preserved omicron-specific functional humoral immunity.
Collapse
Affiliation(s)
| | | | - Paulina Kaplonek
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Massachusetts General Hospital Department of Pediatrics, Mucosal Immunology and Biology Research Center, Boston, MA, USA
- Massachusetts General Hospital Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Vincent Center for Reproductive Biology, Boston, MA, USA
- Massachusetts General Hospital Food Allergy Center, Division of Pediatric Allergy and Immunology, Boston, MA, USA
- Children's Clinical University Hospital, Riga, Latvia
- Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London, UK
| | - Jaewon Kang
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Evan C Lam
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Madeleine D Burns
- Massachusetts General Hospital Department of Pediatrics, Mucosal Immunology and Biology Research Center, Boston, MA, USA
| | - Eva J Farkas
- Massachusetts General Hospital Department of Pediatrics, Mucosal Immunology and Biology Research Center, Boston, MA, USA
| | - Jameson P Davis
- Massachusetts General Hospital Department of Pediatrics, Mucosal Immunology and Biology Research Center, Boston, MA, USA
| | - Brittany P Boribong
- Massachusetts General Hospital Department of Pediatrics, Mucosal Immunology and Biology Research Center, Boston, MA, USA
| | - Andrea G Edlow
- Massachusetts General Hospital Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Vincent Center for Reproductive Biology, Boston, MA, USA
| | - Alessio Fasano
- Massachusetts General Hospital Department of Pediatrics, Mucosal Immunology and Biology Research Center, Boston, MA, USA
| | - Wayne Shreffler
- Massachusetts General Hospital Food Allergy Center, Division of Pediatric Allergy and Immunology, Boston, MA, USA
| | - Dace Zavadska
- Children's Clinical University Hospital, Riga, Latvia
| | - Marina Johnson
- Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London, UK
| | - David Goldblatt
- Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London, UK
| | | | - Lael M Yonker
- Massachusetts General Hospital Department of Pediatrics, Mucosal Immunology and Biology Research Center, Boston, MA, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| |
Collapse
|
10
|
Dick JK, Hart GT. Natural Killer Cell Antibody-Dependent Cellular Cytotoxicity (ADCC) Activity Against Plasmodium falciparum-Infected Red Blood Cells. Methods Mol Biol 2022; 2470:641-657. [PMID: 35881380 DOI: 10.1007/978-1-0716-2189-9_48] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Antibody-dependent cellular cytotoxicity (ADCC) is a mechanism of cell defense that bridges the innate and adaptive immune systems. ADCC has been found to be a major route of immune protection against both viral infections and cancer. Recently, natural killer (NK) cell-mediated ADCC has been shown as a mechanism of parasite clearance and protection against malaria (Hart et al. J Exp Med 216(6):1280-1290, 2019). NK cells bind to antibodies on the surface of Plasmodium falciparum infected red blood cells (iRBCs) via their Fc receptor, FcγRIIIa (CD16). This interaction induces the release of lytic granules (degranulation) by NK cells, which contain perforin, granzyme B, and granulysin, as well as the secretion of IFNγ. ADCC subsequently limits the growth of Plasmodium falciparum in vitro (Arora et al. Elife:7, e36806, 2018). Because NK cell-mediated ADCC has been shown to be important in malaria parasite clearance and protection, further studies understanding ADCC in malaria are warranted (Hart et al. J Exp Med 216(6):1280-1290, 2019). Therefore, this protocol describes methods to perform an in vitro ADCC functional assay with iRBCs. Specifically, it includes protocols on how to grow and culture iRBCs, how to culture peripheral blood mononuclear cells (PMBCs), and how to do in vitro PBMC assays to measure NK degranulation and IFNγ production as measures of NK ADCC function.
Collapse
Affiliation(s)
- Jenna Kelly Dick
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
- Department of Medicine, Division of Infectious Disease and International Medicine, University of Minnesota, Microbiology Research Facility, Minneapolis, MN, USA
- Medical Scientist Training Program, University of Minnesota, Minneapolis, MN, USA
| | - Geoffrey Thomas Hart
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA.
- Department of Medicine, Division of Infectious Disease and International Medicine, University of Minnesota, Microbiology Research Facility, Minneapolis, MN, USA.
| |
Collapse
|
11
|
Nawab DH. Vaccinal antibodies: Fc antibody engineering to improve the antiviral antibody response and induce vaccine-like effects. Hum Vaccin Immunother 2021; 17:5532-5545. [PMID: 34844516 PMCID: PMC8903937 DOI: 10.1080/21645515.2021.1985891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 09/21/2021] [Indexed: 10/19/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic highlights the urgent clinical need for efficient virus therapies and vaccines. Although the functional importance of antibodies is indisputable in viral infections, there are still significant unmet needs that require vast improvements in antibody-based therapeutics. The IgG Fc domain can be engineered to produce antibodies with tailored and potent responses that will meet these clinical demands. Engaging Fc receptors (FcRs) to perform effector functions as cytotoxicity, phagocytosis, complement activation, intracellular neutralization and controlling antibody persistence. Furthermore, it produces vaccine-like effects by activating signals to stimulate T-cell responses, have proven to be required for protection, as neutralization alone does not off the full protection capacity of antibodies. This review highlights antiviral Fc functions and FcRs' contributions in linking innate and adaptive immunity against viral threats. Moreover, it provides the latest Fc engineering strategies to improve the safety and efficacy of human antiviral antibodies and vaccines.
Collapse
Affiliation(s)
- Dhuha H Nawab
- Pharmacy Department, Ministry of Health, Saudi Arabia
| |
Collapse
|
12
|
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
|
13
|
Pourhatami A, Kaviyani-Charati M, Kargar B, Baziyad H, Kargar M, Olmeda-Gómez C. Mapping the intellectual structure of the coronavirus field (2000-2020): a co-word analysis. Scientometrics 2021; 126:6625-6657. [PMID: 34149117 PMCID: PMC8204734 DOI: 10.1007/s11192-021-04038-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 05/08/2021] [Indexed: 12/26/2022]
Abstract
Over the two last decades, coronaviruses have affected human life in different ways, especially in terms of health and economy. Due to the profound effects of novel coronaviruses, growing tides of research are emerging in various research fields. This paper employs a co-word analysis approach to map the intellectual structure of the coronavirus literature for a better understanding of how coronavirus research and the disease itself have developed during the target timeframe. A strategic diagram has been drawn to depict the coronavirus domain's structure and development. A detailed picture of coronavirus literature has been extracted from a huge number of papers to provide a quick overview of the coronavirus literature. The main themes of past coronavirus-related publications are (a) "Antibody-Virus Interactions," (b) "Emerging Infectious Diseases," (c) "Protein Structure-based Drug Design and Antiviral Drug Discovery," (d) "Coronavirus Detection Methods," (e) "Viral Pathogenesis and Immunity," and (f) "Animal Coronaviruses." The emerging infectious diseases are mostly related to fatal diseases (such as Middle East respiratory syndrome, severe acute respiratory syndrome, and COVID-19) and animal coronaviruses (including porcine, turkey, feline, canine, equine, and bovine coronaviruses and infectious bronchitis virus), which are capable of placing animal-dependent industries such as the swine and poultry industries under strong economic pressure. Although considerable research into coronavirus has been done, this unique field has not yet matured sufficiently. Therefore, "Antibody-virus Interactions," "Emerging Infectious Diseases," and "Coronavirus Detection Methods" hold interesting, promising research gaps to be both explored and filled in the future.
Collapse
Affiliation(s)
- Aliakbar Pourhatami
- Department of Information Technology, Faculty of Industrial and Systems Engineering, Tarbiat Modares University, Tehran, Iran
| | | | - Bahareh Kargar
- School of Industrial Engineering, Iran University of Science and Technology, Tehran, Iran
| | - Hamed Baziyad
- Department of Information Technology, Faculty of Industrial and Systems Engineering, Tarbiat Modares University, Tehran, Iran
| | - Maryam Kargar
- School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Carlos Olmeda-Gómez
- Department Library & Information Science, Carlos III University, Madrid, Spain
| |
Collapse
|
14
|
Lamptey H, Bonney EY, Adu B, Kyei GB. Are Fc Gamma Receptor Polymorphisms Important in HIV-1 Infection Outcomes and Latent Reservoir Size? Front Immunol 2021; 12:656894. [PMID: 34017334 PMCID: PMC8129575 DOI: 10.3389/fimmu.2021.656894] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 04/06/2021] [Indexed: 11/13/2022] Open
Abstract
Fc gamma receptors (FcγR) are cell surface glycoproteins which trigger specific effector-cell responses when cross-linked with the Fc portions of immunoglobulin (IgG) antibodies. During HIV-1 infection, the course of disease progression, ART response, and viral reservoir size vary in different individuals. Several factors may account for these differences; however, Fc gamma receptor gene polymorphisms, which influence receptor binding to IgG antibodies, are likely to play a key role. FcγRIIa (CD32) was recently reported as a potential marker for latent HIV reservoir, however, this assertion is still inconclusive. Whether FcγR polymorphisms influence the size of the viral reservoir, remains an important question in HIV cure studies. In addition, potential cure or viral suppression methods such as broadly neutralizing antibody (bNAbs) may depend on FcγRs to control the virus. Here, we discuss the current evidence on the potential role played by FcγR polymorphisms in HIV-1 infection, treatment and vaccine trial outcomes. Importantly, we highlight contrasting findings that may be due to multiple factors and the relatively limited data from African populations. We recommend further studies especially in sub-Saharan Africa to confirm the role of FcγRIIa in the establishment of latent reservoir and to determine their influence in therapies involving bNAbs.
Collapse
Affiliation(s)
- Helena Lamptey
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Evelyn Y. Bonney
- Department of Virology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Bright Adu
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - George B. Kyei
- Department of Virology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
- Department of Medicine, Washington University School of Medicine in St Louis, St. Louis, MO, United States
- Medical and Scientific Research Centre, University of Ghana Medical Centre, University of Ghana, Accra, Ghana
| |
Collapse
|
15
|
Payandeh Z, Mohammadkhani N, Nabi Afjadi M, Khalili S, Rajabibazl M, Houjaghani Z, Dadkhah M. The immunology of SARS-CoV-2 infection, the potential antibody based treatments and vaccination strategies. Expert Rev Anti Infect Ther 2020; 19:899-910. [PMID: 33307883 DOI: 10.1080/14787210.2020.1863144] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Introduction: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) as a potentially fatal agent for a new emerging viral disease (COVID-19) is of great global public health emergency. Herein, we represented potential antibody-based treatments especially monoclonal antibodies (mAbs) that may exert a potential role in treatment as well as developing vaccination strategies against COVID-19.Areas covered: We used PubMed, Google Scholar, and clinicaltrials.gov search strategies for relevant papers. We demonstrated some agents with potentially favorable efficacy as well as favorable safety. Several therapies are under assessment to evaluate their efficacy and safety for COVID19. However, the development of different strategies such as SARS-CoV-2-based vaccines and antibody therapy are urgently required beside other effective therapies such as plasma, anticoagulants, and immune as well as antiviral therapies. We encourage giving more attention to antibody-based treatments as an immediate strategy. Although there has not been any approved specific vaccine until now, developing vaccination strategies may have a protective effect against COVID-19.Expert opinion: An antiviral mAbs could be a safe and high-quality therapeutic intervention which is greatly recommended for COVID-19. Additionally, the high sequence homology between the SARS-CoV-2 and SARS/MERS viruses could shed light on developing to design a vaccine against SARS-CoV-2.
Collapse
Affiliation(s)
- Zahra Payandeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Hospital of Xi'an Jiaotong University (Xibei Hospital), 710004 Xi'an, China
| | - Niloufar Mohammadkhani
- Department of Clinical Biochemistry, School of Medicine, Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohsen Nabi Afjadi
- Institute of Biochemistry and Biophysics, Tehran University, Tehran, Iran
| | - Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran
| | - Masoumeh Rajabibazl
- Department of Clinical Biochemistry, School of Medicine, Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Houjaghani
- Department of Pharmacy Education, EMUPSS, Eastern Mediterranean University, Famagusta, N.Cyprus
| | - Masoomeh Dadkhah
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.,Department of Pharmacology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
16
|
Papasavvas E, Kossenkov AV, Azzoni L, Zetola NM, Mackiewicz A, Ross BN, Fair M, Vadrevu S, Ramogola-Masire D, Sanne I, Firnhaber C, Montaner LJ. Gene expression profiling informs HPV cervical histopathology but not recurrence/relapse after LEEP in ART-suppressed HIV+HPV+ women. Carcinogenesis 2019; 40:225-233. [PMID: 30364933 DOI: 10.1093/carcin/bgy149] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 09/28/2018] [Accepted: 10/24/2018] [Indexed: 12/30/2022] Open
Abstract
Identification of factors associated with human papillomavirus (HPV) cervical histopathology or recurrence/relapse following loop electrosurgical excision procedure (LEEP) would allow for better management of the disease. We investigated whether gene signatures could (i) associate with HPV cervical histopathology and (ii) identify women with post-LEEP disease recurrence/relapse. Gene array analysis was performed on paraffin-embedded cervical tissue-isolated RNA from two cross-sectional cohorts of antiretroviral therapy (ART)-suppressed HIV+HPV+ coinfected women: (i) 55 women in South Africa recruited into three groups: high risk (HR) (-) (n = 16) and HR (+) (n = 15) HPV without cervical histopathology and HR (+) HPV with cervical intraepithelial neoplasia (CIN) grade 1/2/3 (n = 24), (ii) 28 women in Botswana with CIN2/3 treated with LEEP 12-month prior to recruitment and presenting with (n = 13) and without (n = 15) lesion recurrence/relapse (tissue was analyzed at first LEEP). Three distinct gene expression signatures identified were able to segregate: (i) HR+ HPV and CIN1/2/3, (ii) HR HPV-free and cervical histopathology-free and (iii) HR+ HPV and cervical histopathology-free. Immune activation and neoplasia-associated genes (n = 272 genes; e.g. IL-1A, IL-8, TCAM1, POU4F1, MCM2, SMC1B, CXCL6, MMP12) were a feature of cancer precursor dysplasia within HR HPV infection. No difference in LEEP tissue gene expression was detected between women with or without recurrence/relapse. In conclusion, distinctive gene signatures were associated with presence of cervical histopathology in tissues from ART-suppressed HIV+/HPV+ coinfected women. Lack of detection of LEEP tissue gene signature able to segregate subsequent post-LEEP disease recurrence/relapse indicates additional factors independent of local gene expression as determinants of recurrence/relapse.
Collapse
Affiliation(s)
- Emmanouil Papasavvas
- The Wistar Institute, HIV-1 Immunopathogenesis Laboratory, Philadelphia, PA, USA
| | - Andrew V Kossenkov
- The Wistar Institute, HIV-1 Immunopathogenesis Laboratory, Philadelphia, PA, USA
| | - Livio Azzoni
- The Wistar Institute, HIV-1 Immunopathogenesis Laboratory, Philadelphia, PA, USA
| | - Nicola M Zetola
- The Botswana-UPenn Partnership, Department of Radiation Oncology, Gaborone, Botswana.,The University of Pennsylvania, Department of Radiation Oncology, Philadelphia, PA, USA
| | - Agnieszka Mackiewicz
- The Wistar Institute, HIV-1 Immunopathogenesis Laboratory, Philadelphia, PA, USA
| | - Brian N Ross
- The Wistar Institute, HIV-1 Immunopathogenesis Laboratory, Philadelphia, PA, USA
| | - Matthew Fair
- The Wistar Institute, HIV-1 Immunopathogenesis Laboratory, Philadelphia, PA, USA
| | - Surya Vadrevu
- The Wistar Institute, HIV-1 Immunopathogenesis Laboratory, Philadelphia, PA, USA
| | | | - Ian Sanne
- Clinical HIV Research Unit, Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Cynthia Firnhaber
- Clinical HIV Research Unit, Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Right To Care, Johannesburg, South Africa
| | - Luis J Montaner
- The Wistar Institute, HIV-1 Immunopathogenesis Laboratory, Philadelphia, PA, USA
| |
Collapse
|
17
|
Zolla-Pazner S, Gilbert PB. Revisiting the Correlate of Reduced HIV Infection Risk in the Rv144 Vaccine Trial. J Virol 2019; 93:e00629-19. [PMID: 31189712 PMCID: PMC6694814 DOI: 10.1128/jvi.00629-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The RV144 vaccine trial is the only clinical study to have shown a modest but statistically significant decrease in HIV infection risk. RV144 and the subsequent studies identifying the level of V1V2-specific antibodies as a correlate of reduced infection risk are still controversial despite many papers supporting and expanding the initial study. We address these controversies and summarize active-immunization and passive-immunization experiments in nonhuman primates that support the initial finding.
Collapse
Affiliation(s)
- Susan Zolla-Pazner
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Peter B Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center and Department of Biostatistics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
18
|
Geraghty DE, Thorball CW, Fellay J, Thomas R. Effect of Fc Receptor Genetic Diversity on HIV-1 Disease Pathogenesis. Front Immunol 2019; 10:970. [PMID: 31143176 PMCID: PMC6520634 DOI: 10.3389/fimmu.2019.00970] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 04/15/2019] [Indexed: 11/21/2022] Open
Abstract
Fc receptor (FcR) genes collectively have copy number and allelic polymorphisms that have been implicated in multiple inflammatory and autoimmune diseases. This variation might also be involved in etiology of infectious diseases. The protective role of Fc-mediated antibody-function in HIV-1 immunity has led to the investigation of specific polymorphisms in FcR genes on acquisition, disease progression, and vaccine efficacy in natural history cohorts. The purpose of this review is not only to explore these known HIV-1 host genetic associations, but also to re-evaluate them in the context of genome-wide data. In the current era of effective anti-retroviral therapy, the potential impact of such variation on post-treatment cohorts cannot go unheeded and is discussed here in the light of current findings. Specific polymorphisms associating with HIV-1 pathogenesis have previously been genotyped by assays that captured only the single-nucleotide polymorphism (SNP) of interest without relative information of neighboring variants. With recent technological advances, variation within these genes can now be characterized using next-generation sequencing, allowing precise annotation of the whole chromosomal region. We herein also discuss updates in the annotation of common FcR variants that have been previously associated with HIV-1 pathogenesis.
Collapse
Affiliation(s)
- Daniel E Geraghty
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Christian W Thorball
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Jacques Fellay
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.,Precision Medicine Unit, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Rasmi Thomas
- U. S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| |
Collapse
|
19
|
Keshwara R, Shiels T, Postnikova E, Kurup D, Wirblich C, Johnson RF, Schnell MJ. Rabies-based vaccine induces potent immune responses against Nipah virus. NPJ Vaccines 2019; 4:15. [PMID: 31016033 PMCID: PMC6465360 DOI: 10.1038/s41541-019-0109-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 03/04/2019] [Indexed: 12/25/2022] Open
Abstract
Nipah Virus (NiV) is a re-emerging zoonotic pathogen in the genus Henipavirus of the Paramyxoviridae family of viruses. NiV is endemic to Bangladesh and Malaysia and is highly fatal to both livestock and humans (human case fatality rate = 74.5%). Currently, there is no approved vaccine against NiV on the market. The goal of this study was to use a recombinant RABV vector expressing NiV glycoprotein (NiV G) to develop a bivalent candidate vaccine against NiV disease and rabies virus (RABV) disease, which is also a significant health burden in the regions where NiV is endemic. The rabies vector is a well-established vaccine strain that lacks neurovirulence and can stably expresses foreign antigens that are immunogenic in various animal models. Mice inoculated intranasally with the live recombinant RABV/NiV vaccine (NIPARAB) showed no signs of disease. To test the immunogenicity of the vaccine candidate, groups of C57BL/6 mice were immunized intramuscularly with a single dose of live vaccine particles or two doses of chemically inactivated viral particles. Both vaccination groups showed NiV G-specific seroconversion, and the inactivated (INAC) vaccine group yielded higher titers of NiV G-specific antibodies. Furthermore, cross-reactivity of NiV G-specific immune sera against Hendra virus (HeV), was confirmed by immunofluorescence (IF) and indirect ELISA against soluble recombinant HeV glycoprotein (HeV G). Both live and killed vaccines induced neutralizing antibodies. These results indicate that NIPARAB may be used as a killed virus vaccine to protect humans against NiV and RABV, and possibly as a preventative measure against HeV as well.
Collapse
Affiliation(s)
- Rohan Keshwara
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Thomas Shiels
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Elena Postnikova
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, MD 21702 USA
| | - Drishya Kurup
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Christoph Wirblich
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Reed F. Johnson
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - Matthias J. Schnell
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107 USA
- Jefferson Vaccine Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107 USA
| |
Collapse
|
20
|
Williams KL, Stumpf M, Naiman NE, Ding S, Garrett M, Gobillot T, Vézina D, Dusenbury K, Ramadoss NS, Basom R, Kim PS, Finzi A, Overbaugh J. Identification of HIV gp41-specific antibodies that mediate killing of infected cells. PLoS Pathog 2019; 15:e1007572. [PMID: 30779811 PMCID: PMC6396944 DOI: 10.1371/journal.ppat.1007572] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 03/01/2019] [Accepted: 01/08/2019] [Indexed: 11/19/2022] Open
Abstract
Antibodies that mediate killing of HIV-infected cells through antibody-dependent cellular cytotoxicity (ADCC) have been implicated in protection from HIV infection and disease progression. Despite these observations, these types of HIV antibodies are understudied compared to neutralizing antibodies. Here we describe four monoclonal antibodies (mAbs) obtained from one individual that target the HIV transmembrane protein, gp41, and mediate ADCC activity. These four mAbs arose from independent B cell lineages suggesting that in this individual, multiple B cell responses were induced by the gp41 antigen. Competition and phage peptide display mapping experiments suggested that two of the mAbs target epitopes in the cysteine loop that are highly conserved and a common target of HIV gp41-specific antibodies. The amino acid sequences that bind these mAbs are overlapping but distinct. The two other mAbs were competed by mAbs that target the C-terminal heptad repeat (CHR) and the fusion peptide proximal region (FPPR) and appear to both target a similar unique conformational epitope. These gp41-specific mAbs mediated killing of infected cells that express high levels of Env due to either pre-treatment with interferon or deletion of vpu to increase levels of BST-2/Tetherin. They also mediate killing of target cells coated with various forms of the gp41 protein, including full-length gp41, gp41 ectodomain or a mimetic of the gp41 stump. Unlike many ADCC mAbs that target HIV gp120, these gp41-mAbs are not dependent on Env structural changes associated with membrane-bound CD4 interaction. Overall, the characterization of these four new mAbs that target gp41 and mediate ADCC provides evidence for diverse gp41 B cell lineages with overlapping but distinct epitopes within an individual. Such antibodies that can target various forms of envelope protein could represent a common response to a relatively conserved HIV epitope for a vaccine.
Collapse
Affiliation(s)
- Katherine L. Williams
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle WA, United States of America
| | - Megan Stumpf
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle WA, United States of America
| | - Nicole Elise Naiman
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle WA, United States of America
- Molecular and Cellular Biology Graduate Program, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA United States of America
- Medical Scientist Training Program, University of Washington, Seattle WA, United States of America
| | - Shilei Ding
- Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montreal, QC, Canada
| | - Meghan Garrett
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle WA, United States of America
- Molecular and Cellular Biology Graduate Program, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA United States of America
| | - Theodore Gobillot
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle WA, United States of America
- Molecular and Cellular Biology Graduate Program, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA United States of America
- Medical Scientist Training Program, University of Washington, Seattle WA, United States of America
| | - Dani Vézina
- Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montreal, QC, Canada
| | - Katharine Dusenbury
- Medical Scientist Training Program, University of Washington, Seattle WA, United States of America
- Divisions of Basic Sciences and Computational Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
- Department of Genome Sciences, University of Washington, Seattle, WA, United States of America
| | - Nitya S. Ramadoss
- Stanford ChEM-H and Department of Biochemistry, Stanford University, Stanford, CA, United States of America
| | - Ryan Basom
- Genomics and Bioinformatics Shared Resource, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Peter S. Kim
- Stanford ChEM-H and Department of Biochemistry, Stanford University, Stanford, CA, United States of America
- Chan Zuckerberg Biohub, San Francisco, CA, United States of America
| | - Andrés Finzi
- Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montreal, QC, Canada
| | - Julie Overbaugh
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle WA, United States of America
| |
Collapse
|
21
|
Wines BD, Billings H, Mclean MR, Kent SJ, Hogarth PM. Antibody Functional Assays as Measures of Fc Receptor-Mediated Immunity to HIV - New Technologies and their Impact on the HIV Vaccine Field. Curr HIV Res 2018; 15:202-215. [PMID: 28322167 PMCID: PMC5543561 DOI: 10.2174/1570162x15666170320112247] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 02/20/2017] [Accepted: 03/09/2017] [Indexed: 12/23/2022]
Abstract
Background: There is now intense interest in the role of HIV-specific antibodies and the engagement of FcγR functions in the control and prevention of HIV infection. The analyses of the RV144 vaccine trial, natural progression cohorts, and macaque models all point to a role for Fc-dependent effector functions, such as cytotoxicity (ADCC) or phagocytosis (ADCP), in the control of HIV. However, reliable assays that can be reproducibly used across different laboratories to measure Fc-dependent functions, such as antibody dependent cellular cytotoxicity (ADCC) are limited. Method: This brief review highlights the importance of Fc properties for immunity to HIV, particular-ly via FcγR diversity and function. We discuss assays used to study FcR mediated functions of HIV-specific Ab, including our recently developed novel cell-free ELISA using homo-dimeric FcγR ecto-domains to detect functionally relevant viral antigen-specific antibodies. Results: The binding of these dimeric FcγR ectodomains, to closely spaced pairs of IgG Fc, mimics the engagement and cross-linking of Fc receptors by IgG opsonized virions or infected cells as the es-sential prerequisite to the induction of Ab-dependent effector functions. The dimeric FcγR ELISA reli-ably correlates with ADCC in patient responses to influenza. The assay is amenable to high throughput and could be standardized across laboratories. Conclusion: We propose the assay has broader implications for the evaluation of the quality of anti-body responses in viral infections and for the rapid evaluation of responses in vaccine development campaigns for HIV and other viral infections.
Collapse
Affiliation(s)
- Bruce D Wines
- Centre for Biomedical Research, Burnet Institute, Melbourne, Vic 3004, Australia.,Department of Immunology, Monash University Central Clinical School, Melbourne, Victoria 3004, Australia.,Department of Pathology, The University of Melbourne, Victoria, 3010, Australia
| | - Hugh Billings
- Centre for Biomedical Research, Burnet Institute, Melbourne, Vic 3004, Australia
| | - Milla R Mclean
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Parkville, Victoria, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Parkville, Victoria, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Melbourne, Parkville, Victoria, Australia.,Melbourne Sexual Health Centre, Infectious Diseases Department, Alfred Health, Central Clinical School, Monash University, Victoria, Australia
| | - P Mark Hogarth
- Centre for Biomedical Research, Burnet Institute, Melbourne, Vic 3004, Australia.,Department of Immunology, Monash University Central Clinical School, Melbourne, Victoria 3004, Australia.,Department of Pathology, The University of Melbourne, Victoria, 3010, Australia
| |
Collapse
|
22
|
Bissa M, Forlani G, Zanotto C, Tosi G, De Giuli Morghen C, Accolla RS, Radaelli A. Fowlpoxvirus recombinants coding for the CIITA gene increase the expression of endogenous MHC-II and Fowlpox Gag/Pro and Env SIV transgenes. PLoS One 2018; 13:e0190869. [PMID: 29385169 PMCID: PMC5791965 DOI: 10.1371/journal.pone.0190869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 12/21/2017] [Indexed: 01/12/2023] Open
Abstract
A complete eradication of an HIV infection has never been achieved by vaccination and the search for new immunogens that can induce long-lasting protective responses is ongoing. Avipoxvirus recombinants are host-restricted for replication to avian species and they do not have the undesired side effects induced by vaccinia recombinants. In particular, Fowlpox (FP) recombinants can express transgenes over long periods and can induce protective immunity in mammals, mainly due to CD4-dependent CD8+ T cells. In this context, the class II transactivator (CIITA) has a pivotal role in triggering the adaptive immune response through induction of the expression of class-II major histocompatibility complex molecule (MHC-II), that can present antigens to CD4+ T helper cells. Here, we report on construction of novel FPgp and FPenv recombinants that express the highly immunogenic SIV Gag-pro and Env structural antigens. Several FP-based recombinants, with single or dual genes, were also developed that express CIITA, driven from H6 or SP promoters. These recombinants were used to infect CEF and Vero cells in vitro and determine transgene expression, which was evaluated by real-time PCR and Western blotting. Subcellular localisation of the different proteins was evaluated by confocal microscopy, whereas HLA-DR or MHC-II expression was measured by flow cytometry. Fowlpox recombinants were also used to infect syngeneic T/SA tumour cells, then injected into Balb/c mice to elicit MHC-II immune response and define the presentation of the SIV transgene products in the presence or absence of FPCIITA. Antibodies to Env were measured by ELISA. Our data show that the H6 promoter was more efficient than SP to drive CIITA expression and that CIITA can enhance the levels of the gag/pro and env gene products only when infection is performed by FP single recombinants. Also, CIITA expression is higher when carried by FP single recombinants than when combined with FPgp or FPenv constructs and can induce HLA-DR cell surface expression. However, in-vivo experiments did not show any significant increase in the humoral response. As CIITA already proved to elicit immunogenicity by improving antigen presentation, further in-vivo experiments should be performed to increase the immune responses. The use of prime/boost immunisation protocols and the oral administration route of the recombinants may enhance the immunogenicity of Env peptides presented by MHC-II and provide CD4+ T-cell stimulation.
Collapse
Affiliation(s)
- Massimiliano Bissa
- Department of Pharmacological and Biomolecular Sciences, University of Milan, via Balzaretti 9, Milan, Italy
| | - Greta Forlani
- Department of Experimental Medicine, University of Insubria, Via O. Rossi 9, Varese, Italy
| | - Carlo Zanotto
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, via Vanvitelli 32, Milan, Italy
| | - Giovanna Tosi
- Department of Experimental Medicine, University of Insubria, Via O. Rossi 9, Varese, Italy
| | - Carlo De Giuli Morghen
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, via Vanvitelli 32, Milan, Italy
- Catholic University “Our Lady of Good Counsel”, Rr. Dritan Hoxha, Tirana, Albania
| | - Roberto S. Accolla
- Department of Experimental Medicine, University of Insubria, Via O. Rossi 9, Varese, Italy
| | - Antonia Radaelli
- Department of Pharmacological and Biomolecular Sciences, University of Milan, via Balzaretti 9, Milan, Italy
- CNR Institute of Neurosciences, Cellular and Molecular Pharmacology Section, University of Milan, via Vanvitelli 32, Milan, Italy
- * E-mail:
| |
Collapse
|
23
|
Malherbe DC, Mendy J, Vang L, Barnette PT, Reed J, Lakhashe SK, Owuor J, Gach JS, Legasse AW, Axthelm MK, LaBranche CC, Montefiori D, Forthal DN, Park B, Wilson JM, McLinden JH, Xiang J, Stapleton JT, Sacha JB, Haynes BF, Liao HX, Ruprecht RM, Smith J, Gurwith M, Haigwood NL, Alexander J. Combination Adenovirus and Protein Vaccines Prevent Infection or Reduce Viral Burden after Heterologous Clade C Simian-Human Immunodeficiency Virus Mucosal Challenge. J Virol 2018; 92:e01092-17. [PMID: 29093095 PMCID: PMC5752948 DOI: 10.1128/jvi.01092-17] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 10/13/2017] [Indexed: 01/24/2023] Open
Abstract
HIV vaccine development is focused on designing immunogens and delivery methods that elicit protective immunity. We evaluated a combination of adenovirus (Ad) vectors expressing HIV 1086.C (clade C) envelope glycoprotein (Env), SIV Gag p55, and human pegivirus GBV-C E2 glycoprotein. We compared replicating simian (SAd7) with nonreplicating human (Ad4) adenovirus-vectored vaccines paired with recombinant proteins in a novel prime-boost regimen in rhesus macaques, with the goal of eliciting protective immunity against SHIV challenge. In both vaccine groups, plasma and buccal Env-specific IgG, tier 1 heterologous neutralizing antibodies, and antibody-dependent cell-mediated viral inhibition were readily generated. High Env-specific T cell responses elicited in all vaccinees were significantly greater than responses targeting Gag. After three intrarectal exposures to heterologous tier 1 clade C SHIV, all 10 sham-vaccinated controls were infected, whereas 4/10 SAd7- and 3/10 Ad4-vaccinated macaques remained uninfected or maintained tightly controlled plasma viremia. Time to infection was significantly delayed in SAd7-vaccinated macaques compared to the controls. Cell-associated and plasma virus levels were significantly lower in each group of vaccinated macaques compared to controls; the lowest plasma viral burden was found in animals vaccinated with the SAd7 vectors, suggesting superior immunity conferred by the replicating simian vectors. Furthermore, higher V1V2-specific binding antibody titers correlated with viral control in the SAd7 vaccine group. Thus, recombinant Ad plus protein vaccines generated humoral and cellular immunity that was effective in either protecting from SHIV acquisition or significantly reducing viremia in animals that became infected, consequently supporting additional development of replicating Ad vectors as HIV vaccines.IMPORTANCE There is a well-acknowledged need for an effective AIDS vaccine that protects against HIV infection and limits in vivo viral replication and associated pathogenesis. Although replicating virus vectors have been advanced as HIV vaccine platforms, there have not been any direct comparisons of the replicating to the nonreplicating format. The present study directly compared the replicating SAd7 to nonreplicating Ad4 vectors in macaques and demonstrated that in the SAd7 vaccine group, the time to infection was significantly delayed compared to the control group, and V1V2 Env-specific binding antibodies correlated with viral outcomes. Viral control was significantly enhanced in vaccinated macaques compared to controls, and in infected SAd7-vaccinated macaques compared to Ad4-vaccinated macaques, suggesting that this vector may have conferred more effective immunity. Because blocking infection is so difficult with current vaccines, development of a vaccine that can limit viremia if infection occurs would be valuable. These data support further development of replicating adenovirus vectors.
Collapse
Affiliation(s)
- Delphine C Malherbe
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, USA
| | | | - Lo Vang
- PaxVax, Inc., San Diego, California, USA
| | - Philip T Barnette
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Jason Reed
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, USA
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Samir K Lakhashe
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Joshua Owuor
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, Texas, USA
- Southwest National Primate Research Center, San Antonio, Texas, USA
| | - Johannes S Gach
- Division of Infectious Diseases, Department of Medicine, University of California, Irvine School of Medicine, Irvine, California, USA
| | - Alfred W Legasse
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Michael K Axthelm
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, USA
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Celia C LaBranche
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - David Montefiori
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Donald N Forthal
- Division of Infectious Diseases, Department of Medicine, University of California, Irvine School of Medicine, Irvine, California, USA
| | - Byung Park
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, USA
| | - James M Wilson
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James H McLinden
- The Iowa City Veterans Affairs Medical Center, Iowa City, Iowa, USA
- The University of Iowa, Iowa City, Iowa, USA
| | - Jinhua Xiang
- The Iowa City Veterans Affairs Medical Center, Iowa City, Iowa, USA
- The University of Iowa, Iowa City, Iowa, USA
| | - Jack T Stapleton
- The Iowa City Veterans Affairs Medical Center, Iowa City, Iowa, USA
- The University of Iowa, Iowa City, Iowa, USA
| | - Jonah B Sacha
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, USA
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Barton F Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Pathology, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Hua-Xin Liao
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Ruth M Ruprecht
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, Texas, USA
- Southwest National Primate Research Center, San Antonio, Texas, USA
| | | | | | - Nancy L Haigwood
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, USA
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | | |
Collapse
|
24
|
Abstract
Despite major advances in antiretroviral therapy against HIV-1, an effective HIV vaccine is urgently required to reduce the number of new cases of HIV infections in the world. Vaccines are the ultimate tool in the medical arsenal to control and prevent the spread of infectious diseases such as HIV/AIDS. Several failed phase-IIb to –III clinical vaccine trials against HIV-1 in the past generated a plethora of information that could be used for better designing of an effective HIV vaccine in the future. Most of the tested vaccine candidates produced strong humoral responses against the HIV proteins; however, failed to protect due to: 1) the low levels and the narrow breadth of the HIV-1 neutralizing antibodies and the HIV-specific antibody-dependent Fc-mediated effector activities, 2) the low levels and the poor quality of the anti-HIV T-cell responses, and 3) the excessive responses to immunodominant non-protective HIV epitopes, which in some cases blocked the protective immunity and/or enhanced HIV infection. The B-cell epitopes on HIV for producing broadly neutralizing antibodies (bNAbs) against HIV have been extensively characterized, and the next step is to develop bNAb epitope immunogen for HIV vaccine. The bNAb epitopes are often conformational epitopes and therefore more difficult to construct as vaccine immunogen and likely to include immunodominant non-protective HIV epitopes. In comparison, T-cell epitopes are short linear peptides which are easier to construct into vaccine immunogen free of immunodominant non-protective epitopes. However, its difficulty lies in identifying the T-cell epitopes conserved among HIV subtypes and induce long-lasting, potent polyfunctional T-cell and cytotoxic T lymphocyte (CTL) activities against HIV. In addition, these protective T-cell epitopes must be recognized by the HLA prevalent in the country(s) targeted for the vaccine trial. In conclusion, extending from the findings from previous vaccine trials, future vaccines should combine both T- and B-cell epitopes as vaccine immunogen to induce multitude of broad and potent immune effector activities required for sterilizing protection against global HIV subtypes.
Collapse
Affiliation(s)
- Bikash Sahay
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, P.O. Box 110880, Gainesville, FL 32611-0880, USA
| | - Cuong Q Nguyen
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, P.O. Box 110880, Gainesville, FL 32611-0880, USA
| | - Janet K Yamamoto
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, P.O. Box 110880, Gainesville, FL 32611-0880, USA
| |
Collapse
|
25
|
Gao Y, Zhang TY, Yuan Q, Xia NS. Antibody-mediated immunotherapy against chronic hepatitis B virus infection. Hum Vaccin Immunother 2017; 13:1768-1773. [PMID: 28521640 DOI: 10.1080/21645515.2017.1319021] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The currently available drugs to treat hepatitis B virus (HBV) infection include interferons and nucleos(t)ide analogs, which can only induce disease remission and are inefficient for the functional cure of patients with chronic HBV infection (CHB). Since high titers of circulating hepatitis B surface antigen (HBsAg) may be essential to exhaust the host anti-HBV immune response and they cannot be significantly reduced by current drugs, new antiviral strategies aiming to suppress serum hepatitis B surface antigen (HBsAg) could help restore virus-specific immune responses and promote the eradication of the virus. As an alternative strategy, immunotherapy with HBsAg-specific antibodies has shown some direct HBsAg suppression effects in several preclinical and clinical trial studies. However, most described previously HBsAg-specific antibodies only had very short-term HBsAg suppression effects in CHB patients and animal models mimicking persistent HBV infection. More-potent antibodies with long-lasting HBsAg clearance effects are required for the development of the clinical application of antibody-mediated immunotherapy for CHB treatment. Our recent study described a novel mAb E6F6 that targets a unique epitope on HBsAg. It could durably suppress the levels of HBsAg and HBV DNA via Fcγ receptor-dependent phagocytosis in vivo. In this commentary, we summarize the current research progress, including the therapeutic roles and mechanisms of antibody-mediated HBV clearance as well as the epitope-determined therapeutic potency of the antibody. These insights may provide some clues and guidance to facilitate the development of therapeutic antibodies against persistent viral infection.
Collapse
Affiliation(s)
- Ying Gao
- a State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics , School of Public Health, Xiamen University , Xiamen , China.,b National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Science , Xiamen University , Xiamen , China
| | - Tian-Ying Zhang
- a State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics , School of Public Health, Xiamen University , Xiamen , China.,b National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Science , Xiamen University , Xiamen , China
| | - Quan Yuan
- a State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics , School of Public Health, Xiamen University , Xiamen , China.,b National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Science , Xiamen University , Xiamen , China
| | - Ning-Shao Xia
- a State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics , School of Public Health, Xiamen University , Xiamen , China.,b National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Science , Xiamen University , Xiamen , China
| |
Collapse
|
26
|
Lack of ADCC Breadth of Human Nonneutralizing Anti-HIV-1 Antibodies. J Virol 2017; 91:JVI.02440-16. [PMID: 28122982 DOI: 10.1128/jvi.02440-16] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 01/19/2017] [Indexed: 01/23/2023] Open
Abstract
Anti-human immunodeficiency virus type 1 (HIV-1) nonneutralizing antibodies (nnAbs) capable of antibody-dependent cellular cytotoxicity (ADCC) have been identified as a protective immune correlate in the RV144 vaccine efficacy trial. Broadly neutralizing antibodies (bNAbs) also mediate ADCC in cell culture and rely on their Fc region for optimal efficacy in animal models. Here, we selected 9 monoclonal nnAbs and 5 potent bNAbs targeting various epitopes and conformations of the gp120/41 complex and analyzed the potency of the two types of antibodies to bind and eliminate HIV-1-infected cells in culture. Regardless of their neutralizing activity, most of the selected antibodies recognized and killed cells infected with two laboratory-adapted HIV-1 strains. Some nnAbs also bound bystander cells that may have captured viral proteins. However, in contrast to the bNAbs, the nnAbs bound poorly to reactivated infected cells from 8 HIV-positive individuals and did not mediate effective ADCC against these cells. The nnAbs also inefficiently recognize cells infected with 8 different transmitted-founder (T/F) isolates. The addition of a synthetic CD4 mimetic enhanced the binding and killing efficacy of some of the nnAbs in an epitope-dependent manner without reaching the levels achieved by the most potent bNAbs. Overall, our data reveal important qualitative and quantitative differences between nnAbs and bNAbs in their ADCC capacity and strongly suggest that the breadth of recognition of HIV-1 by nnAbs is narrow.IMPORTANCE Most of the anti-HIV antibodies generated by infected individuals do not display potent neutralizing activities. These nonneutralizing antibodies (nnAbs) with antibody-dependent cellular cytotoxicity (ADCC) have been identified as a protective immune correlate in the RV144 vaccine efficacy trial. However, in primate models, the nnAbs do not protect against simian-human immunodeficiency virus (SHIV) acquisition. Thus, the role of nnAbs with ADCC activity in protection from infection remains debatable. In contrast, broadly neutralizing antibodies (bNAbs) neutralize a large array of viral strains and mediate ADCC in cell culture. We analyzed the capacities of 9 nnAbs and 5 bNAbs to eliminate infected cells. We selected 18 HIV-1 strains, including virus reactivated from the reservoir of HIV-positive individuals and transmitted-founder isolates. We report that the nnAbs bind poorly to cells infected with primary HIV-1 strains and do not mediate potent ADCC. Overall, our data show that the breadth of recognition of HIV-1 by nnAbs is narrow.
Collapse
|
27
|
Phage Therapy in Bacterial Infections Treatment: One Hundred Years After the Discovery of Bacteriophages. Curr Microbiol 2016; 74:277-283. [PMID: 27896482 PMCID: PMC5243869 DOI: 10.1007/s00284-016-1166-x] [Citation(s) in RCA: 192] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 11/21/2016] [Indexed: 12/13/2022]
Abstract
The therapeutic use of bacteriophages has seen a renewal of interest blossom in the last few years. This reversion is due to increased difficulties in the treatment of antibiotic-resistant strains of bacteria. Bacterial resistance to antibiotics, a serious problem in contemporary medicine, does not implicate resistance to phage lysis mechanisms. Lytic bacteriophages are able to kill antibiotic-resistant bacteria at the end of the phage infection cycle. Thus, the development of phage therapy is potentially a way to improve the treatment of bacterial infections. However, there are antibacterial phage therapy difficulties specified by broadening the knowledge of the phage nature and influence on the host. It has been shown during experiments that both innate and adaptive immunity are involved in the clearance of phages from the body. Immunological reactions against phages are related to the route of administration and may vary depending on the type of bacterial viruses. For that reason, it is very important to test the immunological response of every single phage, particularly if intravenous therapy is being considered. The lack of these data in previous years was one of the reasons for phage therapy abandonment despite its century-long study. Promising results of recent research led us to look forward to a phage therapy that can be applied on a larger scale and subsequently put it into practice.
Collapse
|
28
|
Rationally Designed Vaccines Targeting the V2 Region of HIV-1 gp120 Induce a Focused, Cross-Clade-Reactive, Biologically Functional Antibody Response. J Virol 2016; 90:10993-11006. [PMID: 27630234 DOI: 10.1128/jvi.01403-16] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 09/04/2016] [Indexed: 01/27/2023] Open
Abstract
Strong antibody (Ab) responses against V1V2 epitopes of the human immunodeficiency virus type 1 (HIV-1) gp120 envelope (Env) correlated with reduced infection rates in studies of HIV, simian-human immunodeficiency virus (SHIV), and simian immunodeficiency virus (SIV). In order to focus the Ab response on V1V2, we used six V1V2 sequences and nine scaffold proteins to construct immunogens which were tested using various immunization regimens for their ability to induce cross-reactive and biologically active V2 Abs in rabbits. A prime/boost immunization strategy was employed using gp120 DNA and various V1V2-scaffold proteins. The rabbit polyclonal Ab responses (i) were successfully focused on the V1V2 region, with weak or only transient responses to other Env epitopes, (ii) displayed broad cross-reactive binding activity with gp120s and the V1V2 regions of diverse strains from clades B, C, and E, (iii) included V2 Abs with specificities similar to those found in HIV-infected individuals, and (iv) remained detectable ≥1 year after the last boosting dose. Importantly, sera from rabbits receiving V1V2-scaffold immunogens displayed Ab-dependent cellular phagocytosis whereas sera from rabbits receiving only gp120 did not. The results represent the first fully successful example of reverse vaccinology in the HIV vaccine field with rationally designed epitope scaffold immunogens inducing Abs that recapitulate the epitope specificity and biologic activity of the human monoclonal Abs from which the immunogens were designed. Moreover, this is the first immunogenicity study using epitope-targeting, rationally designed vaccine constructs that induced an Fc-mediated activity associated with protection from infection with HIV, SIV, and SHIV. IMPORTANCE Novel immunogens were designed to focus the antibody response of rabbits on the V1V2 epitopes of HIV-1 gp120 since such antibodies were associated with reduced infection rates of HIV, SIV, and SHIV. The vaccine-induced antibodies were broadly cross-reactive with the V1V2 regions of HIV subtypes B, C and E and, importantly, facilitated Fc-mediated phagocytosis, an activity not induced upon immunization of rabbits with gp120. This is the first immunogenicity study of vaccine constructs that focuses the antibody response on V1V2 and induces V2-specific antibodies with the ability to mediate phagocytosis, an activity that has been associated with protection from infection with HIV, SIV, and SHIV.
Collapse
|
29
|
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]
|
30
|
Pelegrin M, Naranjo-Gomez M, Piechaczyk M. Antiviral Monoclonal Antibodies: Can They Be More Than Simple Neutralizing Agents? Trends Microbiol 2016; 23:653-665. [PMID: 26433697 PMCID: PMC7127033 DOI: 10.1016/j.tim.2015.07.005] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 07/06/2015] [Accepted: 07/15/2015] [Indexed: 12/12/2022]
Abstract
Monoclonal antibodies (mAbs) are increasingly being considered as agents to fight severe viral diseases. So far, they have essentially been selected and used on the basis of their virus-neutralizing activity and/or cell-killing activity to blunt viral propagation via direct mechanisms. There is, however, accumulating evidence that they can also induce long-lasting protective antiviral immunity by recruiting the endogenous immune system of infected individuals during the period of immunotherapy. Exploiting this property may revolutionize antiviral mAb-based immunotherapies, with benefits for both patients and healthcare systems. Antiviral monoclonal antibodies (mAbs) are promising, high-added-value biotherapeutics. During recent years, the number of antiviral mAbs developed against both acute and chronic viruses has grown exponentially, some of them being currently tested in clinical trials. Antiviral mAbs can be used to blunt viral propagation through direct effects. They can also engage the host's immune system, leading to the induction of long-lasting protective vaccine-like effects. The assessment of mechanisms at play in the induction of vaccine-like effects by antiviral mAbs will help in improving antiviral treatments. Exploiting this effect will translate into therapeutic benefit for patients. The benefit will also help healthcare systems through the reduction of treatment costs.
Collapse
Affiliation(s)
- Mireia Pelegrin
- Equipe Labellisée par la Ligue contre le Cancer - Institut de Génétique Moléculaire de Montpellier, UMR 5535 CNRS, 1919 route de Mende, 34293 Montpellier cedex 5, Université de Montpellier, 163 rue Auguste Broussonnet, 34090 Montpellier, France.
| | - Mar Naranjo-Gomez
- Equipe Labellisée par la Ligue contre le Cancer - Institut de Génétique Moléculaire de Montpellier, UMR 5535 CNRS, 1919 route de Mende, 34293 Montpellier cedex 5, Université de Montpellier, 163 rue Auguste Broussonnet, 34090 Montpellier, France
| | - Marc Piechaczyk
- Equipe Labellisée par la Ligue contre le Cancer - Institut de Génétique Moléculaire de Montpellier, UMR 5535 CNRS, 1919 route de Mende, 34293 Montpellier cedex 5, Université de Montpellier, 163 rue Auguste Broussonnet, 34090 Montpellier, France
| |
Collapse
|
31
|
Duhan V, Khairnar V, Friedrich SK, Zhou F, Gassa A, Honke N, Shaabani N, Gailus N, Botezatu L, Khandanpour C, Dittmer U, Häussinger D, Recher M, Hardt C, Lang PA, Lang KS. Virus-specific antibodies allow viral replication in the marginal zone, thereby promoting CD8(+) T-cell priming and viral control. Sci Rep 2016; 6:19191. [PMID: 26805453 PMCID: PMC4726415 DOI: 10.1038/srep19191] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 11/09/2015] [Indexed: 02/08/2023] Open
Abstract
Clinically used human vaccination aims to induce specific antibodies that can guarantee long-term protection against a pathogen. The reasons that other immune components often fail to induce protective immunity are still debated. Recently we found that enforced viral replication in secondary lymphoid organs is essential for immune activation. In this study we used the lymphocytic choriomeningitis virus (LCMV) to determine whether enforced virus replication occurs in the presence of virus-specific antibodies or virus-specific CD8(+) T cells. We found that after systemic recall infection with LCMV-WE the presence of virus-specific antibodies allowed intracellular replication of virus in the marginal zone of spleen. In contrast, specific antibodies limited viral replication in liver, lung, and kidney. Upon recall infection with the persistent virus strain LCMV-Docile, viral replication in spleen was essential for the priming of CD8(+) T cells and for viral control. In contrast to specific antibodies, memory CD8(+) T cells inhibited viral replication in marginal zone but failed to protect mice from persistent viral infection. We conclude that virus-specific antibodies limit viral infection in peripheral organs but still allow replication of LCMV in the marginal zone, a mechanism that allows immune boosting during recall infection and thereby guarantees control of persistent virus.
Collapse
Affiliation(s)
- Vikas Duhan
- Institute of Immunology of the University Hospital in Essen, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Vishal Khairnar
- Institute of Immunology of the University Hospital in Essen, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Sarah-Kim Friedrich
- Institute of Immunology of the University Hospital in Essen, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Fan Zhou
- Institute of Immunology of the University Hospital in Essen, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Asmae Gassa
- Institute of Immunology of the University Hospital in Essen, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany.,Department of Cardiothoracic Surgery, Cologne University, Heart Center, Kerpener strasse 62, 50937 Cologne, Germany
| | - Nadine Honke
- Institute of Immunology of the University Hospital in Essen, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Namir Shaabani
- Institute of Immunology of the University Hospital in Essen, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Nicole Gailus
- Institute of Immunology of the University Hospital in Essen, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Lacramioara Botezatu
- Department of Hematology of the University Hospital in Essen, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Cyrus Khandanpour
- Department of Hematology of the University Hospital in Essen, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Ulf Dittmer
- Institute of Virology of the University Hospital in Essen, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Dieter Häussinger
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, Düsseldorf 40225, Germany
| | - Mike Recher
- Clinic for Primary Immunodeficiency, Medical Outpatient Unit and Immunodeficiency Laboratory, Department of Biomedicine, University Hospital, Basel, Switzerland
| | - Cornelia Hardt
- Institute of Immunology of the University Hospital in Essen, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Philipp A Lang
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, Düsseldorf 40225, Germany.,Molecular Medicine II, Heinrich-Heine-University, Moorenstrasse 5, Düsseldorf 40225, Germany
| | - Karl S Lang
- Institute of Immunology of the University Hospital in Essen, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany.,Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, Düsseldorf 40225, Germany
| |
Collapse
|
32
|
Gómez RS, Ramirez BA, Céspedes PF, Cautivo KM, Riquelme SA, Prado CE, González PA, Kalergis AM. Contribution of Fcγ receptors to human respiratory syncytial virus pathogenesis and the impairment of T-cell activation by dendritic cells. Immunology 2016; 147:55-72. [PMID: 26451966 PMCID: PMC4693880 DOI: 10.1111/imm.12541] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 09/16/2015] [Accepted: 09/29/2015] [Indexed: 12/11/2022] Open
Abstract
Human respiratory syncytial virus (hRSV) is the leading cause of infant hospitalization related to respiratory disease. Infection with hRSV produces abundant infiltration of immune cells into the airways, which combined with an exacerbated pro-inflammatory immune response can lead to significant damage to the lungs. Human RSV re-infection is extremely frequent, suggesting that this virus may have evolved molecular mechanisms that interfere with host adaptive immunity. Infection with hRSV can be reduced by administering a humanized neutralizing antibody against the virus fusion protein in high-risk infants. Although neutralizing antibodies against hRSV effectively block the infection of airway epithelial cells, here we show that both, bone marrow-derived dendritic cells (DCs) and lung DCs undergo infection with IgG-coated virus (hRSV-IC), albeit abortive. Yet, this is enough to negatively modulate DC function. We observed that such a process is mediated by Fcγ receptors (FcγRs) expressed on the surface of DCs. Remarkably, we also observed that in the absence of hRSV-specific antibodies FcγRIII knockout mice displayed significantly less cellular infiltration in the lungs after hRSV infection, compared with wild-type mice, suggesting a potentially harmful, IgG-independent role for this receptor in hRSV disease. Our findings support the notion that FcγRs can contribute significantly to the modulation of DC function by hRSV and hRSV-IC. Further, we provide evidence for an involvement of FcγRIII in the development of hRSV pathogenesis.
Collapse
MESH Headings
- Adaptive Immunity
- Animals
- Antibodies, Neutralizing/pharmacology
- Antibodies, Viral/immunology
- Antibodies, Viral/metabolism
- Antiviral Agents/pharmacology
- Cells, Cultured
- Coculture Techniques
- Cytokines/metabolism
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dendritic Cells/virology
- Disease Models, Animal
- Immunoglobulin G/immunology
- Immunoglobulin G/metabolism
- Lung/drug effects
- Lung/immunology
- Lung/metabolism
- Lung/virology
- Lymphocyte Activation/drug effects
- Mice, Inbred C57BL
- Mice, Knockout
- Palivizumab/pharmacology
- Receptors, IgG/deficiency
- Receptors, IgG/genetics
- Receptors, IgG/immunology
- Receptors, IgG/metabolism
- Respiratory Syncytial Virus Infections/drug therapy
- Respiratory Syncytial Virus Infections/genetics
- Respiratory Syncytial Virus Infections/immunology
- Respiratory Syncytial Virus Infections/metabolism
- Respiratory Syncytial Virus Infections/virology
- Respiratory Syncytial Virus, Human/drug effects
- Respiratory Syncytial Virus, Human/immunology
- Respiratory Syncytial Virus, Human/pathogenicity
- Signal Transduction
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/virology
- Viral Load
- Virus Replication
Collapse
Affiliation(s)
- Roberto S. Gómez
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiago de ChileChile
| | - Bruno A. Ramirez
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiago de ChileChile
| | - Pablo F. Céspedes
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiago de ChileChile
| | - Kelly M. Cautivo
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiago de ChileChile
| | - Sebastián A. Riquelme
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiago de ChileChile
- INSERM U1064NantesFrance
| | - Carolina E. Prado
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiago de ChileChile
| | - Pablo A. González
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiago de ChileChile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiago de ChileChile
- INSERM U1064NantesFrance
- Departamento de ReumatologíaFacultad de Medicina. Pontificia Universidad Católica de ChileSantiago de ChileChile
| |
Collapse
|
33
|
Nimmerjahn F. A constant threat for HIV: Fc-engineering to enhance broadly neutralizing antibody activity for immunotherapy of the acquired immunodeficiency syndrome. Eur J Immunol 2015; 45:2183-90. [PMID: 26140474 DOI: 10.1002/eji.201445386] [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] [Received: 04/24/2015] [Revised: 06/29/2015] [Accepted: 06/30/2015] [Indexed: 01/12/2023]
Abstract
Passive immunotherapy with polyclonal or hyperimmune serum immunoglobulin G (IgG) preparations provides an efficient means of protecting immunocompromised patients from microbial infections. More recently, the use of passive immunotherapy to prevent or to treat established infections with the human immunodeficiency virus (HIV) has gained much attention, due to promising preclinical data obtained in monkey and humanized mouse in vivo model systems, demonstrating that the transfer of HIV-specific antibodies can not only prevent HIV infection, but also diminish virus load during chronic infection. Furthermore, an array of broadly neutralizing HIV-specific antibodies has become available and the importance of the IgG constant region as a critical modulator of broadly neutralizing activity has been demonstrated. The aim of this review is to summarize the most recent findings with regard to the molecular and cellular mechanisms responsible for antibody-mediated clearance of HIV infection, and to discuss how this may help to improve HIV therapy via optimizing Fcγ-receptor-dependent activities of HIV-specific antibodies.
Collapse
Affiliation(s)
- Falk Nimmerjahn
- Institute of Genetics, Department of Biology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
34
|
Burton DR, Mascola JR. Antibody responses to envelope glycoproteins in HIV-1 infection. Nat Immunol 2015; 16:571-6. [PMID: 25988889 DOI: 10.1038/ni.3158] [Citation(s) in RCA: 324] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 03/26/2015] [Indexed: 02/08/2023]
Abstract
Antibody responses to the HIV-1 envelope glycoproteins can be classified into three groups. Binding but non-neutralizing responses are directed to epitopes that are expressed on isolated envelope glycoproteins but not on the native envelope trimer found on the surface of virions and responsible for mediating the entry of virus into target cells. Strain-specific responses and broadly neutralizing responses, in contrast, target epitopes that are expressed on the native trimer, as revealed by recently resolved structures. The past few years have seen the isolation of many broadly neutralizing antibodies of remarkable potency that have shown prophylactic and therapeutic activities in animal models. These antibodies are helping to guide rational vaccine design and therapeutic strategies for HIV-1.
Collapse
Affiliation(s)
- Dennis R Burton
- 1] Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, USA. [2] International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, California, USA. [3] Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, California, USA. [4] Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Boston, Massachusetts, USA
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
35
|
Feng H, Zhang H, Deng J, Wang L, He Y, Wang S, Seyedtabaei R, Wang Q, Liu L, Galipeau J, Compans RW, Wang BZ. Incorporation of a GPI-anchored engineered cytokine as a molecular adjuvant enhances the immunogenicity of HIV VLPs. Sci Rep 2015; 5:11856. [PMID: 26150163 PMCID: PMC4493578 DOI: 10.1038/srep11856] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 05/22/2015] [Indexed: 12/15/2022] Open
Abstract
HIV vaccines should elicit immune responses at both the mucosal portals of entry to block transmission and systemic compartments to clear disseminated viruses. Co-delivery of mucosal adjuvants has been shown to be essential to induce effective mucosal immunity by non-replicating vaccines. A novel cytokine, GIFT4, engineered by fusing GM-CSF and interleukin-4, was previously found to simulate B cell proliferation and effector function. Herein a membrane-anchored form of GIFT4 was constructed by fusing a glycolipid (GPI)-anchoring sequence and incorporated into Env-enriched HIV virus-like particles (VLPs) as a molecular adjuvant. Guinea pigs were immunized with the resulting HIV VLPs through an intramuscular priming-intranasal boosting immunization route. The GIFT4-containing VLPs induced higher levels of systemic antibody responses with significantly increased binding avidity and improved neutralizing breadth and potency to a panel of selected strains, as well as higher levels of IgG and IgA at several mucosal sites. Thus, the novel GPI-GIFT4-containging VLPs have the potential to be developed into a prophylactic HIV vaccine. Incorporation of GPI-anchored GIFT4 into VLPs as a molecular adjuvant represents a novel approach to increase their immunogenicity.
Collapse
Affiliation(s)
- Hao Feng
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA
| | - Han Zhang
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA
| | - Jiusheng Deng
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | - Li Wang
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA
| | - Yuan He
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA
| | - Shelly Wang
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA
| | - Roheila Seyedtabaei
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA
| | - Qing Wang
- Department of Bioengineering, Henan University of Technology, Zhengzhou 450052, China
| | - Laiting Liu
- Department of Bioengineering, Henan University of Technology, Zhengzhou 450052, China
| | - Jacques Galipeau
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | - Richard W Compans
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA
| | - Bao-Zhong Wang
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
36
|
Nimmerjahn F, Gordan S, Lux A. FcγR dependent mechanisms of cytotoxic, agonistic, and neutralizing antibody activities. Trends Immunol 2015; 36:325-36. [PMID: 25981969 DOI: 10.1016/j.it.2015.04.005] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 04/16/2015] [Accepted: 04/16/2015] [Indexed: 12/21/2022]
Abstract
Given the widespread use of antibodies of the immunoglobulin G (IgG) class as cytotoxic, immunomodulatory, and neutralizing agents in the therapy of malignant, infectious, and autoimmune diseases, understanding the molecular and cellular mechanisms responsible for their therapeutic activity is of major importance. While Fcγ receptors (FcγR) have well-appreciated roles as effectors of cytotoxic IgG activity, it has only recently become clear that the functionality of immunomodulatory and neutralizing IgG preparations also depends on cellular FcγRs. Here, we review current models of IgG activity in infectious and inflammatory settings, and examine the importance of cell type-specific expression of FcγRs in determining functional outcome. We discuss how this knowledge may be used to improve the activity of therapeutic antibody preparations and outline important areas of focus for future research.
Collapse
Affiliation(s)
- Falk Nimmerjahn
- Institute of Genetics at the Department of Biology, Friedrich-Alexander University Erlangen-Nürnberg, Erwin-Rommelstrasse 3, 91058 Erlangen, Germany.
| | - Sina Gordan
- Institute of Genetics at the Department of Biology, Friedrich-Alexander University Erlangen-Nürnberg, Erwin-Rommelstrasse 3, 91058 Erlangen, Germany
| | - Anja Lux
- Institute of Genetics at the Department of Biology, Friedrich-Alexander University Erlangen-Nürnberg, Erwin-Rommelstrasse 3, 91058 Erlangen, Germany
| |
Collapse
|
37
|
Dotsey EY, Gorlani A, Ingale S, Achenbach CJ, Forthal DN, Felgner PL, Gach JS. A High Throughput Protein Microarray Approach to Classify HIV Monoclonal Antibodies and Variant Antigens. PLoS One 2015; 10:e0125581. [PMID: 25938510 PMCID: PMC4418728 DOI: 10.1371/journal.pone.0125581] [Citation(s) in RCA: 13] [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: 12/22/2014] [Accepted: 03/24/2015] [Indexed: 11/19/2022] Open
Abstract
In recent years, high throughput discovery of human recombinant monoclonal antibodies (mAbs) has been applied to greatly advance our understanding of the specificity, and functional activity of antibodies against HIV. Thousands of antibodies have been generated and screened in functional neutralization assays, and antibodies associated with cross-strain neutralization and passive protection in primates, have been identified. To facilitate this type of discovery, a high throughput-screening tool is needed to accurately classify mAbs, and their antigen targets. In this study, we analyzed and evaluated a prototype microarray chip comprised of the HIV-1 recombinant proteins gp140, gp120, gp41, and several membrane proximal external region peptides. The protein microarray analysis of 11 HIV-1 envelope-specific mAbs revealed diverse binding affinities and specificities across clades. Half maximal effective concentrations, generated by our chip analysis, correlated significantly (P<0.0001) with concentrations from ELISA binding measurements. Polyclonal immune responses in plasma samples from HIV-1 infected subjects exhibited different binding patterns, and reactivity against printed proteins. Examining the totality of the specificity of the humoral response in this way reveals the exquisite diversity, and specificity of the humoral response to HIV.
Collapse
Affiliation(s)
- Emmanuel Y. Dotsey
- Division of Infectious Diseases, University of California Irvine, Irvine, California, United States of America
| | - Andrea Gorlani
- Division of Infectious Diseases, University of California Irvine, Irvine, California, United States of America
| | - Sampat Ingale
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Chad J. Achenbach
- Division of Infectious Diseases, Northwestern University, Chicago, Illinois, United States of America
| | - Donald N. Forthal
- Division of Infectious Diseases, University of California Irvine, Irvine, California, United States of America
| | - Philip L. Felgner
- Division of Infectious Diseases, University of California Irvine, Irvine, California, United States of America
- * E-mail: (JSG); (PLF)
| | - Johannes S. Gach
- Division of Infectious Diseases, University of California Irvine, Irvine, California, United States of America
- * E-mail: (JSG); (PLF)
| |
Collapse
|
38
|
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
|
39
|
Zhou J, Amran FS, Kramski M, Angelovich TA, Elliott J, Hearps AC, Price P, Jaworowski A. An NK Cell Population Lacking FcRγ Is Expanded in Chronically Infected HIV Patients. THE JOURNAL OF IMMUNOLOGY 2015; 194:4688-97. [PMID: 25855354 DOI: 10.4049/jimmunol.1402448] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 03/06/2015] [Indexed: 01/17/2023]
Abstract
We previously demonstrated that NK cells from HIV-infected individuals have elevated expression of activation markers, spontaneously degranulate ex vivo, and decrease expression of a signal-transducing protein for NK-activating receptors, FcRγ. Importantly, these changes were maintained in virologically suppressed (VS) individuals receiving combination antiretroviral therapy (cART). In this study, we show that loss of FcRγ is caused by the expansion of a novel subset of FcRγ(-)CD56(dim) NK cells with an altered activation receptor repertoire and biological properties. In a cross-sectional study, FcRγ(-) NK cells as a proportion of total CD56(dim) NK cells increased in cART-naive viremic HIV-infected individuals (median [interquartile range] = 25.9 [12.6-56.1] compared with 3.80 [1.15-11.5] for HIV(-) controls, p < 0.0001) and in VS HIV-infected individuals (22.7 [13.1-56.2] compared with 3.80 [1.15-11.5], p = 0.0004), with no difference between cART-naive and VS patients (p = 0.93). FcRγ(-) NK cells expressed no NKp30 or NKp46. They showed greater Ab-dependent cellular cytotoxicity activity against rituximab-opsonized Raji cells and in a whole-blood assay measuring NK responses to overlapping HIV peptides, despite having reduced CD16 expression compared with conventional NK cells. Their prevalence correlated with CMV Ab titers in HIV(-) subjects but not in HIV(+) individuals, and with the inflammatory marker CXCL10 in both groups. The expansion of a subset of NK cells that lacks NKp30 and NKp46 to ∼90% of CD56(dim) NK cells in some VS HIV(+) individuals may influence NK-mediated immunosurveillance in patients receiving cART.
Collapse
Affiliation(s)
- Jingling Zhou
- Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria 3004, Australia
| | - Fathiah S Amran
- School of Pathology and Laboratory Medicine, University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Marit Kramski
- Department of Immunology and Microbiology, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Tom A Angelovich
- Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria 3004, Australia; School of Applied Sciences, Royal Melbourne Institute of Technology University, Melbourne, Victoria 3000, Australia
| | - Julian Elliott
- Department of Infectious Diseases, Monash University, Melbourne, Victoria 3004, Australia; and
| | - Anna C Hearps
- Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria 3004, Australia; Department of Infectious Diseases, Monash University, Melbourne, Victoria 3004, Australia; and
| | - Patricia Price
- School of Pathology and Laboratory Medicine, University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Anthony Jaworowski
- Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria 3004, Australia; Department of Infectious Diseases, Monash University, Melbourne, Victoria 3004, Australia; and Department of Immunology, Monash University, Melbourne, Victoria 3004, Australia
| |
Collapse
|
40
|
Weis JF, McClelland RS, Jaoko W, Mandaliya KN, Overbaugh J, Graham SM. Short communication: Fc gamma receptors IIa and IIIa genetic polymorphisms do not predict HIV-1 disease progression in Kenyan women. AIDS Res Hum Retroviruses 2015; 31:288-92. [PMID: 25312792 DOI: 10.1089/aid.2014.0209] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Genetic polymorphisms of the Fc gamma receptors (FcγR) IIa and IIIa have been implicated in the rate of HIV-1 disease progression, but results are inconsistent. We aimed to determine the association between these polymorphisms and disease progression in a cohort of HIV-1 seroconverters from Mombasa, Kenya. Neither FcγRIIa nor FcγRIIIa genotypes were predictive of set point viral load, viral load increase, CD4 decline, or HIV-1 disease progression (time to CD4 count <200 cells/mm(3), death, or treatment initiation). Our results suggest that FcγR polymorphisms might not be an important indicator of viral control and disease progression in this population.
Collapse
Affiliation(s)
- Julie F. Weis
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Institute for Public Health Genetics, University of Washington, Seattle, Washington
| | - R. Scott McClelland
- Department of Medicine, University of Washington, Seattle, Washington
- Department of Epidemiology, University of Washington, Seattle, Washington
- Department of Global Health, University of Washington, Seattle, Washington
- Institute of Tropical and Infectious Diseases, University of Nairobi, Nairobi, Kenya
| | - Walter Jaoko
- Department of Medical Microbiology, University of Nairobi, Nairobi, Kenya
| | | | - Julie Overbaugh
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Susan M. Graham
- Department of Medicine, University of Washington, Seattle, Washington
- Department of Epidemiology, University of Washington, Seattle, Washington
- Department of Global Health, University of Washington, Seattle, Washington
- Institute of Tropical and Infectious Diseases, University of Nairobi, Nairobi, Kenya
| |
Collapse
|
41
|
Passive transfer of neutralizing mAb KD-247 reduces plasma viral load in patients chronically infected with HIV-1. AIDS 2015; 29:453-62. [PMID: 25630040 DOI: 10.1097/qad.0000000000000570] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Neutralizing antibodies against HIV-1 such as a humanized mAb KD-247 can mediate effector functions that attack infected cells in vitro. However, the clinical efficacy of neutralizing antibodies in infected individuals remains to be determined. We evaluated the safety, tolerability and pharmacokinetics of KD-247 infusion and its effect on plasma HIV-1 RNA load and CD4 T-cell count. DESIGN AND METHODS KD-1002 is a phase Ib, double-blind, placebo-controlled, dose-escalation study of KD-247 in asymptomatic HIV-1 seropositive individuals who did not need antiretroviral therapy. Individuals were randomized to 4, 8 or 16 mg/kg KD-247 or placebo, and received three infusions over a 2-week period. RESULTS Patients were randomized to receive one of the three doses of KD-247 and the treatment was well tolerated. We observed a significant decrease in HIV RNA in the 8 and 16 mg/kg KD-247 cohorts, with two individuals who achieved more than 1 log reduction of HIV RNA. Two patients in the 16 mg/kg cohort had selections and/or mutations in the V3-tip region that suggested evasion of neutralization. Long-term suppression of viral load was observed in one patient despite a significant decrease in plasma concentration of KD-247, suggesting effects of the antibody other than neutralization or loss of fitness of the evading virus. CONCLUSION The results indicate that KD-247 reduces viral load in patients with chronic HIV-1 infection and further clinical trials are warranted.
Collapse
|
42
|
Receptor binding domain based HIV vaccines. BIOMED RESEARCH INTERNATIONAL 2015; 2015:594109. [PMID: 25667925 PMCID: PMC4312573 DOI: 10.1155/2015/594109] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 10/16/2014] [Indexed: 11/17/2022]
Abstract
This paper analyzes the main trend of the development of acquired immunodeficiency syndrome (AIDS) vaccines in recent years. Designing an HIV-1 vaccine that provides robust protection from HIV-1 infection remains a challenge despite many years of effort. Therefore, we describe the receptor binding domain of gp120 as a target for developing AIDS vaccines. And we recommend some measures that could induce efficiently and produce cross-reactive neutralizing antibodies with high binding affinity. Those measures may offer a new way of the research and development of the potent and broad AIDS vaccines.
Collapse
|
43
|
Abstract
ABSTRACT
Antibodies can impact pathogens in the presence or in the absence of effector cells or effector molecules such as complement, and experiments can often sort out with precision the mechanisms by which an antibody inhibits a pathogen
in vitro
. In addition,
in vivo
models, particularly those engineered to knock in or knock out effector cells or effector molecules, are excellent tools for understanding antibody functions. However, it is highly likely that multiple antibody functions occur simultaneously or sequentially in the presence of an infecting organism
in vivo
. The most critical incentive for measuring antibody functions is to provide a basis for vaccine development and for the development of therapeutic antibodies. In this respect, some functions, such as virus neutralization, serve to inhibit the acquisition of a pathogen or limit its pathogenesis. However, antibodies can also enhance replication or contribute to pathogenesis. This review emphasizes those antibody functions that are potentially beneficial to the host. In addition, this review will focus on the effects of antibodies on organisms themselves, rather than on the toxins the organisms may produce.
Collapse
|
44
|
Forthal DN. Functions of Antibodies. Microbiol Spectr 2014; 2:1-17. [PMID: 25215264 PMCID: PMC4159104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023] Open
Affiliation(s)
- Donald N. Forthal
- Chief, Infectious Diseases, University of California, Irvine, 3044 Hewitt Hall, Irvine, CA 92617, 949-824-3366
| |
Collapse
|
45
|
Lema D, Garcia A, De Sanctis JB. HIV vaccines: a brief overview. Scand J Immunol 2014; 80:1-11. [PMID: 24813074 DOI: 10.1111/sji.12184] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 04/22/2014] [Indexed: 02/06/2023]
Abstract
The scope of the article is to review the different approaches that have been used for HIV vaccines. The review is based on articles retrieved by PubMed and clinical trials from 1990 up to date. The article discusses virus complexity, protective and non-protective immune responses against the virus, and the most important approaches for HIV vaccine development.
Collapse
Affiliation(s)
- D Lema
- Instituto de Inmunología, Facultad de Medicina, Universidad Central de Venezuela, Caracas, Venezuela
| | | | | |
Collapse
|
46
|
Basu D, Xiao P, Ende Z, Bere A, Britt WJ, Mulenga J, Kilembe W, Allen SA, Derdeyn CA, Hunter E. Low antibody-dependent cellular cytotoxicity responses in Zambians prior to HIV-1 intrasubtype C superinfection. Virology 2014; 462-463:295-8. [PMID: 25004405 PMCID: PMC4125417 DOI: 10.1016/j.virol.2014.06.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 06/07/2014] [Accepted: 06/15/2014] [Indexed: 11/17/2022]
Abstract
We have previously shown that HIV-1 superinfected Zambian seroconverters mount low binding and neutralizing antibody responses to their primary HIV-1 infecting virus, which could increase susceptibility to re-infection. Here, we investigated if antibody-dependent cellular cytotoxicity (ADCC), a process by which virus-infected cells are killed, was also reduced. Superinfected individuals exhibited low ADCC activity compared to non-superinfected individuals, but similar levels of CMV-reactive binding antibodies, suggesting superinfected individuals are capable of generating and maintaining virus-specific antibodies.
Collapse
Affiliation(s)
- Debby Basu
- Emory Vaccine Center at Yerkes National Primate Research Center, 954 Gatewood Road, Atlanta, GA 30329, USA.
| | - Peng Xiao
- Emory Vaccine Center at Yerkes National Primate Research Center, 954 Gatewood Road, Atlanta, GA 30329, USA.
| | - Zachary Ende
- Emory Vaccine Center at Yerkes National Primate Research Center, 954 Gatewood Road, Atlanta, GA 30329, USA.
| | - Alfred Bere
- Emory Vaccine Center at Yerkes National Primate Research Center, 954 Gatewood Road, Atlanta, GA 30329, USA.
| | - William J Britt
- University of Alabama - Birmingham, 1600 7th Avenue South, Children׳s Harbor Building 107, Birmingham, AL 35233-1711, USA.
| | - Joseph Mulenga
- Zambia Emory HIV Research Project (ZEHRP), B22/737 Mwembelelo, Emmasdale, Post Net 412; P/BagE891, Zambia.
| | - William Kilembe
- Zambia Emory HIV Research Project (ZEHRP), B22/737 Mwembelelo, Emmasdale, Post Net 412; P/BagE891, Zambia.
| | - Susan A Allen
- Zambia Emory HIV Research Project (ZEHRP), B22/737 Mwembelelo, Emmasdale, Post Net 412; P/BagE891, Zambia; Department of Global Health, Rollins School of Public Health, Emory University, 1518 Clifton Rd NE, Atlanta, GA 30322, USA; Department of Pathology and Laboratory Medicine, Emory University, 101, Woodruff Circle, Atlanta, GA 30322, USA.
| | - Cynthia A Derdeyn
- Emory Vaccine Center at Yerkes National Primate Research Center, 954 Gatewood Road, Atlanta, GA 30329, USA; Department of Pathology and Laboratory Medicine, Emory University, 101, Woodruff Circle, Atlanta, GA 30322, USA.
| | - Eric Hunter
- Emory Vaccine Center at Yerkes National Primate Research Center, 954 Gatewood Road, Atlanta, GA 30329, USA; Department of Pathology and Laboratory Medicine, Emory University, 101, Woodruff Circle, Atlanta, GA 30322, USA.
| |
Collapse
|
47
|
Morales JF, Morin TJ, Yu B, Tatsuno GP, O'Rourke SM, Theolis R, Mesa KA, Berman PW. HIV-1 envelope proteins and V1/V2 domain scaffolds with mannose-5 to improve the magnitude and quality of protective antibody responses to HIV-1. J Biol Chem 2014; 289:20526-42. [PMID: 24872420 PMCID: PMC4110267 DOI: 10.1074/jbc.m114.554089] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 05/07/2014] [Indexed: 01/15/2023] Open
Abstract
Two lines of investigation have highlighted the importance of antibodies to the V1/V2 domain of gp120 in providing protection from HIV-1 infection. First, the recent RV144 HIV-1 vaccine trial documented a correlation between non-neutralizing antibodies to the V2 domain and protection. Second, multiple broadly neutralizing monoclonal antibodies to the V1/V2 domain (e.g. PG9) have been isolated from rare infected individuals, termed elite neutralizers. Interestingly, the binding of both types of antibodies appears to depend on the same cluster of amino acids (positions 167–171) adjacent to the junction of the B and C strands of the four-stranded V1/V2 domain β-sheet structure. However, the broadly neutralizing mAb, PG9, additionally depends on mannose-5 glycans at positions 156 and 160 for binding. Because the gp120 vaccine immunogens used in previous HIV-1 vaccine trials were enriched for complex sialic acid-containing glycans, and lacked the high mannose structures required for the binding of PG9-like mAbs, we wondered if these immunogens could be improved by limiting glycosylation to mannose-5 glycans. Here, we describe the PG9 binding activity of monomeric gp120s from multiple strains of HIV-1 produced with mannose-5 glycans. We also describe the properties of glycopeptide scaffolds from the V1/V2 domain also expressed with mannose-5 glycans. The V1/V2 scaffold from the A244 isolate was able to bind the PG9, CH01, and CH03 mAbs with high affinity provided that the proper glycans were present. We further show that immunization with A244 V1/V2 fragments alone, or in a prime/boost regimen with gp120, enhanced the antibody response to sequences in the V1/V2 domain associated with protection in the RV144 trial.
Collapse
Affiliation(s)
- Javier F. Morales
- From the Department of Biomolecular Engineering, Baskin School of Engineering, University of California at Santa Cruz, Santa Cruz, California 95064
| | - Trevor J. Morin
- From the Department of Biomolecular Engineering, Baskin School of Engineering, University of California at Santa Cruz, Santa Cruz, California 95064
| | - Bin Yu
- From the Department of Biomolecular Engineering, Baskin School of Engineering, University of California at Santa Cruz, Santa Cruz, California 95064
| | - Gwen P. Tatsuno
- From the Department of Biomolecular Engineering, Baskin School of Engineering, University of California at Santa Cruz, Santa Cruz, California 95064
| | - Sara M. O'Rourke
- From the Department of Biomolecular Engineering, Baskin School of Engineering, University of California at Santa Cruz, Santa Cruz, California 95064
| | - Richard Theolis
- From the Department of Biomolecular Engineering, Baskin School of Engineering, University of California at Santa Cruz, Santa Cruz, California 95064
| | - Kathryn A. Mesa
- From the Department of Biomolecular Engineering, Baskin School of Engineering, University of California at Santa Cruz, Santa Cruz, California 95064
| | - Phillip W. Berman
- From the Department of Biomolecular Engineering, Baskin School of Engineering, University of California at Santa Cruz, Santa Cruz, California 95064
| |
Collapse
|
48
|
Su B, Moog C. Which Antibody Functions are Important for an HIV Vaccine? Front Immunol 2014; 5:289. [PMID: 24995008 PMCID: PMC4062070 DOI: 10.3389/fimmu.2014.00289] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 06/03/2014] [Indexed: 01/18/2023] Open
Abstract
HIV antibody (Ab) functions capable of preventing mucosal cell-free or cell-to-cell HIV transmission are critical for the development of effective prophylactic and therapeutic vaccines. In addition to CD4(+) T cells, other potential HIV-target cell types including antigen-presenting cells (APCs) (dendritic cells, macrophages) residing at mucosal sites are infected. Moreover, the interactions between APCs and HIV lead to HIV cell-to-cell transmission. Recently discovered broadly neutralizing antibodies (NAbs) are able to neutralize a broad spectrum of HIV strains, inhibit cell-to-cell transfer, and efficiently protect from infection in the experimentally challenged macaque model. However, the 31% protection observed in the RV144 vaccine trial in the absence of detectable NAbs in blood samples pointed to the possible role of additional Ab inhibitory functions. Increasing evidence suggests that IgG Fcγ receptor (FcγR)-mediated inhibition of Abs present at the mucosal site may play a role in protection against HIV mucosal transmission. Moreover, mucosal IgA Abs may be determinant in protection against HIV sexual transmission. Therefore, defining Ab inhibitory functions that could lead to protection is critical for further HIV vaccine design. Here, we review different inhibitory properties of HIV-specific Abs and discuss their potential role in protection against HIV sexual transmission.
Collapse
Affiliation(s)
- Bin Su
- INSERM U1109, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg , Strasbourg , France
| | - Christiane Moog
- INSERM U1109, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg , Strasbourg , France
| |
Collapse
|
49
|
HIV‐specific antibody‐dependent phagocytosis matures during HIV infection. Immunol Cell Biol 2014; 92:679-87. [DOI: 10.1038/icb.2014.42] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 04/29/2014] [Accepted: 04/29/2014] [Indexed: 02/02/2023]
|
50
|
Pollara J, Bonsignori M, Moody MA, Pazgier M, Haynes BF, Ferrari G. Epitope specificity of human immunodeficiency virus-1 antibody dependent cellular cytotoxicity [ADCC] responses. Curr HIV Res 2014; 11:378-87. [PMID: 24191939 PMCID: PMC3878369 DOI: 10.2174/1570162x113116660059] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 09/19/2013] [Accepted: 09/28/2013] [Indexed: 12/02/2022]
Abstract
Antibody dependent cellular cytotoxicity [ADCC] has been suggested to play an important role in control of Human Immunodeficiency Virus-1 [HIV-1] viral load and protection from infection. ADCC antibody responses have been mapped to multiple linear and conformational epitopes within the HIV-1 envelope glycoproteins gp120 and gp41. Many epitopes targeted by antibodies that mediate ADCC overlap with those recognized by antibodies capable of virus neutralization. In addition, recent studies conducted with human monoclonal antibodies derived from HIV-1 infected individuals and HIV-1 vaccine-candidate vaccinees have identified a number of antibodies that lack the ability to capture primary HIV-1 isolates or mediate neutralizing activity, but are able to bind to the surface of infected CD4+ T cells and mediate ADCC. Of note, the conformational changes in the gp120 that may not exclusively relate to binding of the CD4 molecule are important in exposing epitopes recognized by ADCC responses. Here we discuss the HIV-1 envelope epitopes targeted by ADCC antibodies in the context of the potential protective capacities of ADCC.
Collapse
Affiliation(s)
- Justin Pollara
- Department of Surgery, Duke University Medical Center, P.O. Box 2926, Durham, NC 27710, USA.
| | | | | | | | | | | |
Collapse
|