1
|
Pinto-Santini D, Jalil EM, Fernandes GT, Hilaire G, Kolevic L, Cabello R, Grinsztejn B, Pape W, Deschamps MM, House MG, Brofsky E, Sahasrabuddhe VV, Dasgupta S, Pasalar S, Madeleine MM, Carter J, Prabhu PR, Galloway D, Duerr A. ULACNet-301, OPTIMO protocol: optimizing HPV vaccination regimen for cancer prevention in children and adolescents living with HIV. BMC Cancer 2025; 25:151. [PMID: 39871186 PMCID: PMC11771066 DOI: 10.1186/s12885-025-13551-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 01/17/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Persistent infection with human papillomavirus (HPV) is associated with most cervical and anal cancer cases and a large fraction of other anogenital and oropharyngeal cancers. The prophylactic HPV vaccines are known to prevent HPV infections and HPV-associated disease, although there is evidence of reduced response to the HPV vaccination among individuals living with HIV. Prior studies among individuals without HIV suggest that a single HPV vaccine dose induces humoral immune responses that, while lower than those induced by two or three doses, still confer protection against HPV infection. Current recommendations for HPV vaccine include a single-dose schedule for children 9-14-years-olds without HIV. Although two to three doses are recommended for children living with HIV (CLWH), there is very limited data comparing responses to one vs. 2-3 doses in CLWH. METHODS The OPTIMO study will compare immune responses to HPV vaccination in CLWH by measuring antibody and memory B cell (Bmem) responses after 1, 2, or 3 doses of the 9-valent HPV (9vHPV) vaccine, Gardasil-9. A comparison group of children without HIV will receive one dose of the vaccine. The durability of the response will be assessed at 24 months after the last dose of a given regimen. The OPTIMO trial will take place among CLWH from low and middle-income country (LMIC) settings in Peru, Brazil, and Haiti. DISCUSSION Previous studies of single-dose regimens in individuals without HIV raise questions about whether one dose would suffice for CLWH and, if not, whether two or three doses are needed to provide protection against HPV-related cancers. These questions have operational consequences in LMICs given the barriers to delivering multiple doses, uneven availability, and intermittent shortages of HPV vaccines. In addition, information on HIV status for children and adolescents is rarely available during vaccination campaigns based in schools or public health clinics, so CLWH may receive a single dose despite policy recommendations that they receive two or three. This study will provide evidence on the optimal number of doses needed for CLWH that can inform HPV vaccination campaigns in LMICs, especially those with a higher burden of HIV infection and higher incidence of HPV-related cancers. TRIAL REGISTRATION ClinicalTrials.gov NCT04265950.
Collapse
Affiliation(s)
| | - Emilia M Jalil
- Instituto Nacional de Infectología Evandro Chagas-Fiocruz, Rio de Janeiro, Brazil
| | | | - Genevieve Hilaire
- Groupe Haitien d'Etudes de Sarcome de Kaposi et Infections Opportunistes (GHESKIO), Port au Prince, Haiti
| | | | | | - Beatriz Grinsztejn
- Instituto Nacional de Infectología Evandro Chagas-Fiocruz, Rio de Janeiro, Brazil
| | - William Pape
- Groupe Haitien d'Etudes de Sarcome de Kaposi et Infections Opportunistes (GHESKIO), Port au Prince, Haiti
| | - Marie Marcelle Deschamps
- Groupe Haitien d'Etudes de Sarcome de Kaposi et Infections Opportunistes (GHESKIO), Port au Prince, Haiti
| | - Margaret G House
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Emma Brofsky
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Vikrant V Sahasrabuddhe
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | | | | | | | | | | | | | - Ann Duerr
- Fred Hutchinson Cancer Center, Seattle, WA, USA.
| |
Collapse
|
2
|
Qu W, Hua C, Wang Y, Wang Y, Zhang L, Wang Z, Shi W, Chen F, Wu Z, Wang Q, Lu L, Jiang S, Sui L, Li Y. Lineage Replacement and Genetic Changes of Four HR-HPV Types during the Period of Vaccine Coverage: A Six-Year Retrospective Study in Eastern China. Vaccines (Basel) 2024; 12:411. [PMID: 38675793 PMCID: PMC11053858 DOI: 10.3390/vaccines12040411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/31/2024] [Accepted: 04/07/2024] [Indexed: 04/28/2024] Open
Abstract
OBJECTIVE This study aimed to provide clinical evidence for lineage replacement and genetic changes of High-Risk Human Papillomavirus (HR-HPV) during the period of vaccine coverage and characterize those changes in eastern China. METHODS This study consisted of two stages. A total of 90,583 patients visiting the Obstetrics and Gynecology Hospital of Fudan University from March 2018 to March 2022 were included in the HPV typing analysis. Another 1076 patients who tested positive for HPV31, 33, 52, or 58 from November 2020 to August 2023 were further included for HPV sequencing. Vaccination records, especially vaccine types and the third dose administration time, medical history, and cervical cytology samples were collected. Viral DNA sequencing was then conducted, followed by phylogenetic analysis and sequence alignment. RESULTS The overall proportion of HPV31 and 58 infections increased by 1.23% and 0.51%, respectively, while infection by HPV33 and 52 decreased by 0.42% and 1.43%, respectively, within the four-year vaccination coverage period. The proportion of HPV31 C lineage infections showed a 22.17% increase in the vaccinated group, while that of the HPV58 A2 sublineage showed a 12.96% increase. T267A and T274N in the F-G loop of HPV31 L1 protein, L150F in the D-E loop, and T375N in the H-I loop of HPV58 L1 protein were identified as high-frequency escape-related mutations. CONCLUSIONS Differences in epidemic lineage changes and dominant mutation accumulation may result in a proportional difference in trends of HPV infection. New epidemic lineages and high-frequency escape-related mutations should be noted during the vaccine coverage period, and regional epidemic variants should be considered during the development of next-generation vaccines.
Collapse
Affiliation(s)
- Wenjie Qu
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (W.Q.)
| | - Chen Hua
- Shanghai Institute of Infection Disease and Biosecurity, Shanghai Medical College, Fudan University, Shanghai 200032, China; (C.H.)
| | - Yaping Wang
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (W.Q.)
| | - Yan Wang
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (W.Q.)
- Qingpu Branch of the Yangtze River Delta Integrated Demonstration Zone, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200090, China
| | - Lu Zhang
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (W.Q.)
| | - Zhiheng Wang
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (W.Q.)
- Qingpu Branch of the Yangtze River Delta Integrated Demonstration Zone, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200090, China
| | - Wenqian Shi
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (W.Q.)
| | - Fang Chen
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (W.Q.)
| | - Zhiyong Wu
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (W.Q.)
- Qingpu Branch of the Yangtze River Delta Integrated Demonstration Zone, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200090, China
| | - Qian Wang
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, Shanghai 201508, China
| | - Lu Lu
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, Shanghai 201508, China
| | - Shibo Jiang
- Shanghai Institute of Infection Disease and Biosecurity, Shanghai Medical College, Fudan University, Shanghai 200032, China; (C.H.)
| | - Long Sui
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (W.Q.)
| | - Yanyun Li
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (W.Q.)
- Qingpu Branch of the Yangtze River Delta Integrated Demonstration Zone, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200090, China
| |
Collapse
|
3
|
Budeus B, Kibler A, Küppers R. Human IgM-expressing memory B cells. Front Immunol 2023; 14:1308378. [PMID: 38143767 PMCID: PMC10748387 DOI: 10.3389/fimmu.2023.1308378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/27/2023] [Indexed: 12/26/2023] Open
Abstract
A hallmark of T cell dependent (TD) humoral immune responses is the generation of long-lived memory B cells. The generation of these cells occurs primarily in the germinal center (GC) reaction, where antigen-activated B cells undergo affinity maturation as a major consequence of the combined processes of proliferation, somatic hypermutation of their immunoglobulin V (IgV) region genes, and selection for improved affinity of their B-cell antigen receptors. As many B cells also undergo class-switching to IgG or IgA in these TD responses, there was traditionally a focus on class-switched memory B cells in both murine and human studies on memory B cells. However, it has become clear that there is also a large subset of IgM-expressing memory B cells, which have important phenotypic and functional similarities but also differences to class-switched memory B cells. There is an ongoing discussion about the origin of distinct subsets of human IgM+ B cells with somatically mutated IgV genes. We argue here that the vast majority of human IgM-expressing B cells with somatically mutated IgV genes in adults is indeed derived from GC reactions, even though a generation of some mostly lowly mutated IgM+ B cells from other differentiation pathways, mainly in early life, may exist.
Collapse
Affiliation(s)
| | | | - Ralf Küppers
- Institute of Cell Biology (Cancer Research), Medical Faculty, University of Duisburg–Essen, Essen, Germany
| |
Collapse
|
4
|
Qu W, Sui L, Li Y. Vaccine escape challenges virus prevention: The example of two vaccine-preventable oncogenic viruses. J Med Virol 2023; 95:e29184. [PMID: 37943176 DOI: 10.1002/jmv.29184] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/06/2023] [Accepted: 10/09/2023] [Indexed: 11/10/2023]
Abstract
Over the years, the pace of developing vaccines for HBV and HPV has never stopped. After more than 30 years of application, the HBV vaccine has reduced 80% of hepatocellular carcinoma (HCC). However, vaccine escape variants occur under selective pressure induced by widespread vaccination and antiviral therapy, which results in fulminant infection and horizontal transmission. Several mechanisms have been studied to explain HBV vaccine escape, including vaccine escape mutations (VEMs) in the major hydrophilic region, which leads to a decrease in the binding ability to neutralize antibodies and is the primary escape mechanism, protein conformational and N-linked glycosylation sites changes caused by VEMs, differences in genotype distribution, gene recombination, and some temporarily unknown reasons. However, effective solutions are still being explored. The HPV vaccine has also been proven to prevent 70%-90% of cervical cancer worldwide. Cases of HPV infection after being vaccinated have been observed in clinical practice. However, few researchers have paid attention to the mechanism of HPV vaccine escape. Thus, we reviewed the literature on vaccine escape of both HBV and HPV to discuss the mechanism of the virus escaping from vaccine protection and possible solutions to this problem. We analyzed the gap between studies of HPV and HBV and made prospects for further research in HPV vaccine escape.
Collapse
Affiliation(s)
- Wenjie Qu
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China
| | - Long Sui
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Yanyun Li
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|
5
|
Sun Y, Khan MAAK, Mangiola S, Barrow AD. IL17RB and IL17REL Expression Are Associated with Improved Prognosis in HPV-Infected Head and Neck Squamous Cell Carcinomas. Pathogens 2023; 12:pathogens12040572. [PMID: 37111458 PMCID: PMC10143491 DOI: 10.3390/pathogens12040572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/16/2023] [Accepted: 03/27/2023] [Indexed: 04/29/2023] Open
Abstract
Changes in the cellular secretome are implicated in virus infection, malignancy, and anti-tumor immunity. We analyzed the association between transcriptional signatures (TS) from 24 different immune and stromal cell types on the prognosis of HPV-infected and HPV-free head and neck squamous carcinoma (HNSCC) patients from The Cancer Genome Atlas (TCGA) cohort. We found that HPV-positive HNSCC patients have tumors with elevated immune cell TS and improved prognosis, which was specifically associated with an increased tumor abundance of memory B and activated natural killer (NK) cell TS, compared to HPV-free HNSCC patients. HPV-infected patients upregulated many transcripts encoding secreted factors, such as growth factors, hormones, chemokines and cytokines, and their cognate receptors. Analysis of secretome transcripts and cognate receptors revealed that tumor expression of IL17RB and IL17REL are associated with a higher viral load and memory B and activated NK cell TS, as well as improved prognosis in HPV-infected HNSCC patients. The transcriptional parameters that we describe may be optimized to improve prognosis and risk stratification in the clinic and provide insights into gene and cellular targets that may potentially enhance anti-tumor immunity mediated by NK cells and memory B cells in HPV-infected HNSCC patients.
Collapse
Affiliation(s)
- Yuhan Sun
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne 3000, Australia
| | - Md Abdullah Al Kamran Khan
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne 3000, Australia
| | - Stefano Mangiola
- Division of Bioinformatics, Walter and Eliza Hall Institute, Parkville 3052, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne 3010, Australia
| | - Alexander David Barrow
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne 3000, Australia
| |
Collapse
|
6
|
Guenthoer J, Lilly M, Starr TN, Dadonaite B, Lovendahl KN, Croft JT, Stoddard CI, Chohan V, Ding S, Ruiz F, Kopp MS, Finzi A, Bloom JD, Chu HY, Lee KK, Overbaugh J. Identification of broad, potent antibodies to functionally constrained regions of SARS-CoV-2 spike following a breakthrough infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.12.15.520606. [PMID: 36561191 PMCID: PMC9774213 DOI: 10.1101/2022.12.15.520606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The antiviral benefit of antibodies can be compromised by viral escape especially for rapidly evolving viruses. Therefore, durable, effective antibodies must be both broad and potent to counter newly emerging, diverse strains. Discovery of such antibodies is critically important for SARS-CoV-2 as the global emergence of new variants of concern (VOC) has compromised the efficacy of therapeutic antibodies and vaccines. We describe a collection of broad and potent neutralizing monoclonal antibodies (mAbs) isolated from an individual who experienced a breakthrough infection with the Delta VOC. Four mAbs potently neutralize the Wuhan-Hu-1 vaccine strain, the Delta VOC, and also retain potency against the Omicron VOCs, including recently circulating BA.4/BA.5, in both pseudovirus-based and live virus assays, and one also potently neutralizes SARS-CoV-1. The potency of these mAbs was greater against Omicron VOCs than all but one of the mAbs that had been approved for therapeutic applications. The mAbs target distinct epitopes on the spike glycoprotein, three in the receptor binding domain (RBD) and one in an invariant region downstream of the RBD in subdomain 1 (SD1). The escape pathways we defined at single amino acid resolution with deep mutational scanning show they target conserved, functionally constrained regions of the glycoprotein, suggesting escape could incur a fitness cost. Overall, these mAbs are novel in their breadth across VOCs, their epitope specificity, and include a highly potent mAb targeting a rare epitope outside of the RBD in SD1.
Collapse
|
7
|
Structural characterization of protective non-neutralizing antibodies targeting Crimean-Congo hemorrhagic fever virus. Nat Commun 2022; 13:7298. [PMID: 36435827 PMCID: PMC9701186 DOI: 10.1038/s41467-022-34923-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 11/10/2022] [Indexed: 11/28/2022] Open
Abstract
Crimean-Congo Hemorrhagic Fever Virus (CCHFV) causes a life-threatening disease with up to a 40% mortality rate. With no approved medical countermeasures, CCHFV is considered a public health priority agent. The non-neutralizing mouse monoclonal antibody (mAb) 13G8 targets CCHFV glycoprotein GP38 and protects mice from lethal CCHFV challenge when administered prophylactically or therapeutically. Here, we reveal the structures of GP38 bound with a human chimeric 13G8 mAb and a newly isolated CC5-17 mAb from a human survivor. These mAbs bind overlapping epitopes with a shifted angle. The broad-spectrum potential of c13G8 and CC5-17 and the practicality of using them against Aigai virus, a closely related nairovirus were examined. Binding studies demonstrate that the presence of non-conserved amino acids in Aigai virus corresponding region prevent CCHFV mAbs from binding Aigai virus GP38. This information, coupled with in vivo efficacy, paves the way for future mAb therapeutics effective against a wide swath of CCHFV strains.
Collapse
|
8
|
Prabhu PR, Carter JJ, Galloway DA. B Cell Responses upon Human Papillomavirus (HPV) Infection and Vaccination. Vaccines (Basel) 2022; 10:vaccines10060837. [PMID: 35746445 PMCID: PMC9229470 DOI: 10.3390/vaccines10060837] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 02/01/2023] Open
Abstract
Infection with human papillomavirus (HPV) is the necessary cause of cervical cancer. Availability of vaccines against HPV makes it a highly preventable disease. HPV vaccines act through type-specific neutralizing antibodies produced by antigen-specific plasma cells known as long-lived plasma cells (LLPC). However, just as any other vaccine, success of HPV vaccine is attributed to the immunologic memory that it builds, which is largely attained through generation and maintenance of a class of B cells named memory B cells (Bmem). Both LLPCs and Bmems are important in inducing and maintaining immune memory and it is therefore necessary to understand their role after HPV vaccination to better predict outcomes. This review summarizes current knowledge of B-cell responses following HPV vaccination and natural infection, including molecular signatures associated with these responses.
Collapse
|
9
|
Complementary Roles of Antibody Heavy and Light Chain Somatic Hypermutation in Conferring Breadth and Potency to the HIV-1-Specific CAP256-VRC26 bNAb Lineage. J Virol 2022; 96:e0027022. [PMID: 35510865 DOI: 10.1128/jvi.00270-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Some HIV-infected people develop broadly neutralizing antibodies (bNAbs) that block many diverse, unrelated strains of HIV from infecting target cells and, through passive immunization, protect animals and humans from infection. Therefore, understanding the development of bNAbs and their neutralization can inform the design of an HIV vaccine. Here, we extend our previous studies of the ontogeny of the CAP256-VRC26 V2-targeting bNAb lineage by defining the mutations that confer neutralization to the unmutated common ancestor (CAP256.UCA). Analysis of the sequence of the CAP256.UCA showed that many improbable mutations were located in the third complementarity-determining region of the heavy chain (CDRH3) and the heavy chain framework 3 (FR3). Transferring the CDRH3 from bNAb CAP256.25 (63% breadth and 0.003 μg/mL potency) into the CAP256.UCA introduced breadth and the ability to neutralize emerging viral variants. In addition, we showed that the framework and light chain contributed to potency and that the second CDR of the light chain forms part of the paratope of CAP256.25. Notably, a minimally mutated CAP256 antibody, with 41% of the mutations compared to bNAb CAP256.25, was broader (64% breadth) and more potent (0.39 μg/mL geometric potency) than many unrelated bNAbs. Together, we have identified key regions and mutations that confer breadth and potency in a V2-specific bNAb lineage. These data indicate that immunogens that target affinity maturation to key sites in CAP256-VRC26-like precursors, including the CDRHs and light chain, could rapidly elicit breadth through vaccination. IMPORTANCE A major focus in the search for an HIV vaccine is elucidating the ontogeny of broadly neutralizing antibodies (bNAbs), which prevent HIV infection in vitro and in vivo. The unmutated common ancestors (UCAs) of bNAbs are generally strain specific and acquire breadth through extensive, and sometimes redundant, somatic hypermutation during affinity maturation. We investigated which mutations in the CAP256-VRC26 bNAb lineage conferred neutralization capacity to the UCA. We found that mutations in the antibody heavy and light chains had complementary roles in neutralization breadth and potency, respectively. The heavy chain, particularly the third complementarity-determining region, was responsible for conferring breadth. In addition, previously uninvestigated mutations in the framework also contributed to breadth. Together, approximately half of the mutations in CAP256.25 were necessary for broader and more potent neutralization than many unrelated neutralizing antibodies. Vaccine approaches that promote affinity maturation at key sites could therefore more rapidly produce antibodies with neutralization breadth.
Collapse
|
10
|
Shipley MM, Mangala Prasad V, Doepker LE, Dingens A, Ralph DK, Harkins E, Dhar A, Arenz D, Chohan V, Weight H, Mandaliya K, Bloom JD, Matsen FA, Lee KK, Overbaugh JM. Functional development of a V3/glycan-specific broadly neutralizing antibody isolated from a case of HIV superinfection. eLife 2021; 10:68110. [PMID: 34263727 PMCID: PMC8376252 DOI: 10.7554/elife.68110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 07/14/2021] [Indexed: 12/12/2022] Open
Abstract
Stimulating broadly neutralizing antibodies (bnAbs) directly from germline remains a barrier for HIV vaccines. HIV superinfection elicits bnAbs more frequently than single infection, providing clues of how to elicit such responses. We used longitudinal antibody sequencing and structural studies to characterize bnAb development from a superinfection case. BnAb QA013.2 bound initial and superinfecting viral Env, despite its probable naive progenitor only recognizing the superinfecting strain, suggesting both viruses influenced this lineage. A 4.15 Å cryo-EM structure of QA013.2 bound to native-like trimer showed recognition of V3 signatures (N301/N332 and GDIR). QA013.2 relies less on CDRH3 and more on framework and CDRH1 for affinity and breadth compared to other V3/glycan-specific bnAbs. Antigenic profiling revealed that viral escape was achieved by changes in the structurally-defined epitope and by mutations in V1. These results highlight shared and novel properties of QA013.2 relative to other V3/glycan-specific bnAbs in the setting of sequential, diverse antigens.
Collapse
Affiliation(s)
- Mackenzie M Shipley
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Vidya Mangala Prasad
- Department of Medicinal Chemistry, University of Washington, Seattle, United States
| | - Laura E Doepker
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Adam Dingens
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Duncan K Ralph
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Elias Harkins
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Amrit Dhar
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Dana Arenz
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Vrasha Chohan
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Haidyn Weight
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Kishor Mandaliya
- Coast Provincial General Hospital, Women's Health Project, Mombasa, Kenya
| | - Jesse D Bloom
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States.,Department of Genome Sciences, University of Washington, Seattle, United States.,Howard Hughes Medical Institute, Chevy Chase, United States
| | - Frederick A Matsen
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Kelly K Lee
- Department of Medicinal Chemistry, University of Washington, Seattle, United States
| | - Julie M Overbaugh
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, United States
| |
Collapse
|
11
|
Effective high-throughput isolation of fully human antibodies targeting infectious pathogens. Nat Protoc 2021; 16:3639-3671. [PMID: 34035500 DOI: 10.1038/s41596-021-00554-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 04/12/2021] [Indexed: 02/04/2023]
Abstract
As exemplified by the ongoing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic, there is a strong demand for rapid high-throughput isolation pipelines to identify potent neutralizing antibodies for prevention and therapy of infectious diseases. However, despite substantial progress and extensive efforts, the identification and production of antigen-specific antibodies remains labor- and cost-intensive. We have advanced existing concepts to develop a highly efficient high-throughput protocol with proven application for the isolation of potent antigen-specific antibodies against human immunodeficiency virus 1, hepatitis C virus, human cytomegalovirus, Middle East respiratory syndrome coronavirus, SARS-CoV-2 and Ebola virus. It is based on computationally optimized multiplex primer sets (openPrimeR), which guarantee high coverage of even highly mutated immunoglobulin gene segments as well as on optimized antibody cloning and production strategies. Here, we provide the detailed protocol, which covers all critical steps from sample collection to antibody production within 12-14 d.
Collapse
|
12
|
Simonich C, Shipley MM, Doepker L, Gobillot T, Garrett M, Cale EM, Hennessy B, Itell H, Chohan V, Doria-Rose N, Nduati R, Overbaugh J. A diverse collection of B cells responded to HIV infection in infant BG505. Cell Rep Med 2021; 2:100314. [PMID: 34195680 PMCID: PMC8233660 DOI: 10.1016/j.xcrm.2021.100314] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 02/23/2021] [Accepted: 05/18/2021] [Indexed: 12/03/2022]
Abstract
Increasing evidence suggests infants develop unique neutralizing antibody (nAb) responses to HIV compared to adults. Here, we dissected the nAb response of an infant whose virus is in clinical trials as a vaccine immunogen, with a goal of characterizing the broad responses in the infant to this antigen. We isolated 73 nAbs from infant BG505 and identified a large number of clonal families. Twenty-six antibodies neutralized tier 2 viruses-in some cases, viruses from the same clade as BG505, and in others, a different clade, although none showed notable breadth. Several nAbs demonstrated antibody-dependent cellular cytotoxicity activity and targeted the V3 loop. These findings suggest an impressive polyclonal response to HIV infection in infant BG505, adding to the growing evidence that the nAb response to HIV in infants is polyclonal-a desirable vaccine response to a rapidly evolving virus like HIV.
Collapse
Affiliation(s)
- Cassandra Simonich
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA, USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, USA
| | - Mackenzie M. Shipley
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Laura Doepker
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Theodore Gobillot
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA, USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, USA
| | - Meghan Garrett
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, USA
| | - Evan M. Cale
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Brianna Hennessy
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Hannah Itell
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, USA
| | - Vrasha Chohan
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Nicole Doria-Rose
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ruth Nduati
- Department of Pediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Julie Overbaugh
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
13
|
Whaley RE, Ameny S, Arkatkar T, Seese A, Wall A, Khan I, Carter JJ, Scherer EM, Rawlings DJ, Galloway DA, McElrath MJ, Cohen KW, McGuire AT. Generation of a cost-effective cell line for support of high-throughput isolation of primary human B cells and monoclonal neutralizing antibodies. J Immunol Methods 2020; 488:112901. [PMID: 33069767 PMCID: PMC7560121 DOI: 10.1016/j.jim.2020.112901] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 10/09/2020] [Accepted: 10/12/2020] [Indexed: 01/11/2023]
Abstract
The isolation of human monoclonal antibodies (mAbs) arising from natural infection with human pathogens has proven to be a powerful technology, facilitating the understanding of the host response to infection at a molecular level. mAbs can reveal sites of vulnerability on pathogens and illuminate the biological function of the antigenic targets. Moreover, mAbs have the potential to be used directly for therapeutic applications such as passive delivery to prevent infection in susceptible target populations, and as treatment of established infection. The isolation of antigen-specific B cells from vaccine trials can also assist in deciphering whether the desired B cells are being targeted by a given vaccine. Several different processes have been developed to isolate mAbs, but all are generally labor-intensive and result in varying degrees of efficiency. Here, we describe the development of a cost-effective feeder cell line that stably expresses CD40-ligand, interleukin-2 and interleukin-21. Sorting of single B cells onto a layer of irradiated feeder cells sustained antibody production that permits functional screening of secreted antibodies in a manner that enables subsequent recovery of B cells for recombinant antibody cloning. As a proof of concept, we show that this approach can be used to isolate B cells that secrete antibodies that neutralize human papilloma virus (HPV) from participants of an HPV vaccine study. Development of a cell line that provides signals for human B cell growth and antibody secretion. 50% cultures seeded from single B cells produce detectable IgG. B cells secreting HPV-neutralizing antibodies from a vaccine study are identified.
Collapse
Affiliation(s)
- Rachael E Whaley
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Sarah Ameny
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Tanvi Arkatkar
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Aaron Seese
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Abigail Wall
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Iram Khan
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA, USA
| | - Joseph J Carter
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Erin M Scherer
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - David J Rawlings
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, WA, USA; Departments of Pediatrics and Immunology, University of Washington, Seattle, WA, USA
| | - Denise A Galloway
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - M Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Department of Medicine, University of Washington, Seattle, WA, USA
| | - Kristen W Cohen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
| | - Andrew T McGuire
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Department of Global Health, University of Washington, Seattle, WA, USA; Deparment of Laboratory Medicine and Pahthology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
14
|
Isolation of Monoclonal Antibodies from Zika Virus-Infected Patient Samples. Methods Mol Biol 2020; 2142:261-288. [PMID: 32367373 PMCID: PMC7197385 DOI: 10.1007/978-1-0716-0581-3_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
The combination of sorting antigen-specific memory B cells with determining immunoglobulin (Ig) genes at the single-cell level enables the isolation of monoclonal antibodies (mAbs) in individuals. This method requires a small amount of blood (usually 10 mL) and is rapid (less than 2 weeks to isolate antigen-specific mAbs). Due to the application of antigens as the bait to capture the specific memory B cells, the majority of isolated mAbs are true binders to the antigen, which increases the isolation efficiency. Here, applying this approach, we describe the characterization of mAbs against Zika virus from a convalescent patient sample. From 10 mL whole blood, we sorted 33 Zika envelope (E) protein-interacting single memory B cells. The Ig genes from 15 cells were determined, and 13 mAbs were found that bind to Zika E protein with varied binding affinities.
Collapse
|
15
|
Cox AL. Challenges and Promise of a Hepatitis C Virus Vaccine. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a036947. [PMID: 31548228 DOI: 10.1101/cshperspect.a036947] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
An estimated 1.5-2 million new hepatitis C virus (HCV) infections occur globally each year. Critical to the World Health Organization's (WHO) HCV elimination strategy is an 80% reduction in incidence of HCV infections by 2030. However, even among high-income countries, few are on target to achieve the WHO's incident infection-reduction goal. A preventative vaccine could have a major impact in achieving incidence-reduction targets globally. However, barriers to HCV vaccine development are significant and include at-risk populations that are often marginalized: viral diversity, limited options for testing HCV vaccines, and an incomplete understanding of protective immune responses. In part because of those factors, testing of only one vaccine strategy has been completed in at-risk individuals as of 2019. Despite challenges, immunity against HCV protects against chronic infection in some repeated HCV exposures and an effective HCV vaccine could prevent transmission regardless of risk factors. Ultimately, prophylactic vaccines will likely be necessary to achieve global HCV elimination.
Collapse
Affiliation(s)
- Andrea L Cox
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University, Baltimore, Maryland 21205, USA
| |
Collapse
|
16
|
B cell memory: building two walls of protection against pathogens. Nat Rev Immunol 2019; 20:229-238. [PMID: 31836872 PMCID: PMC7223087 DOI: 10.1038/s41577-019-0244-2] [Citation(s) in RCA: 329] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2019] [Indexed: 12/24/2022]
Abstract
Surviving a single infection often results in lifelong immunity to the infecting pathogen. Such protection is mediated, in large part, by two main B cell memory ‘walls’ — namely, long-lived plasma cells and memory B cells. The cellular and molecular processes that drive the production of long-lived plasma cells and memory B cells are subjects of intensive research and have important implications for global health. Indeed, although nearly all vaccines in use today depend on their ability to induce B cell memory, we have not yet succeeded in developing vaccines for some of the world’s most deadly diseases, including AIDS and malaria. Here, we describe the two-phase process by which antigen drives the generation of long-lived plasma cells and memory B cells and highlight the challenges for successful vaccine development in each phase. The authors discuss the formation of two main ‘walls’ of B cell memory to protect against pathogen reinfection. The first wall comprises high-affinity antibodies produced by long-lived plasma cells, while the second wall is formed by memory B cells.
Collapse
|
17
|
Dibo M, Battocchio EC, dos Santos Souza LM, da Silva MDV, Banin-Hirata BK, Sapla MM, Marinello P, Rocha SP, Faccin-Galhardi LC. Antibody Therapy for the Control of Viral Diseases: An Update. Curr Pharm Biotechnol 2019; 20:1108-1121. [DOI: 10.2174/1389201020666190809112704] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 04/22/2019] [Accepted: 08/01/2019] [Indexed: 12/29/2022]
Abstract
The epidemiological impact of viral diseases, combined with the emergence and reemergence of some viruses, and the difficulties in identifying effective therapies, have encouraged several studies to develop new therapeutic strategies for viral infections. In this context, the use of immunotherapy for the treatment of viral diseases is increasing. One of the strategies of immunotherapy is the use of antibodies, particularly the monoclonal antibodies (mAbs) and multi-specific antibodies, which bind directly to the viral antigen and bring about activation of the immune system. With current advancements in science and technology, several such antibodies are being tested, and some are already approved and are undergoing clinical trials. The present work aims to review the status of mAb development for the treatment of viral diseases.
Collapse
Affiliation(s)
- Miriam Dibo
- Department of Microbiology, Biological Sciences Center, State University of Londrina, Parana, Brazil
| | - Eduardo C. Battocchio
- Department of Microbiology, Biological Sciences Center, State University of Londrina, Parana, Brazil
| | - Lucas M. dos Santos Souza
- Department of Microbiology, Biological Sciences Center, State University of Londrina, Parana, Brazil
| | | | - Bruna K. Banin-Hirata
- Department of Pathological Sciences, Biological Sciences Center, State University of Londrina, Parana, Brazil
| | - Milena M.M. Sapla
- Department of Pathological Sciences, Biological Sciences Center, State University of Londrina, Parana, Brazil
| | - Poliana Marinello
- Department of Pathological Sciences, Biological Sciences Center, State University of Londrina, Parana, Brazil
| | - Sérgio P.D. Rocha
- Department of Microbiology, Biological Sciences Center, State University of Londrina, Parana, Brazil
| | - Lígia C. Faccin-Galhardi
- Department of Microbiology, Biological Sciences Center, State University of Londrina, Parana, Brazil
| |
Collapse
|
18
|
Puronen CE, Ford ES, Uldrick TS. Immunotherapy in People With HIV and Cancer. Front Immunol 2019; 10:2060. [PMID: 31555284 PMCID: PMC6722204 DOI: 10.3389/fimmu.2019.02060] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 08/15/2019] [Indexed: 12/16/2022] Open
Abstract
HIV infection alters the natural history of several cancers, in large part due to its effect on the immune system. Immune function in people living with HIV may vary from normal to highly dysfunctional and is largely dependent on the timing of initiation (and continuation) of effective antiretroviral therapy (ART). An individual's level of immune function in turn affects their cancer risk, management, and outcomes. HIV-associated lymphocytopenia and immune dysregulation permit immune evasion of oncogenic viruses and premalignant lesions and are associated with inferior outcomes in people with established cancers. Various types of immunotherapy, including monoclonal antibodies, interferon, cytokines, immunomodulatory drugs, allogeneic hematopoietic stem cell transplant, and most importantly ART have shown efficacy in HIV-related cancer. Emerging data suggest that checkpoint inhibitors targeting the PD-1/PD-L1 pathway can be safe and effective in people with HIV and cancer. Furthermore, some cancer immunotherapies may also affect HIV persistence by influencing HIV latency and HIV-specific immunity. Studying immunotherapy in people with HIV and cancer will advance clinical care of all people living with HIV and presents a unique opportunity to gain insight into mechanisms for HIV eradication.
Collapse
Affiliation(s)
- Camille E Puronen
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Emily S Ford
- Division of Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA, United States.,Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Thomas S Uldrick
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, United States.,Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States.,Division of Global Oncology, Department of Medicine, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| |
Collapse
|
19
|
Boonyaratanakornkit J, Taylor JJ. Techniques to Study Antigen-Specific B Cell Responses. Front Immunol 2019; 10:1694. [PMID: 31396218 PMCID: PMC6667631 DOI: 10.3389/fimmu.2019.01694] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/08/2019] [Indexed: 12/13/2022] Open
Abstract
Antibodies against foreign antigens are a critical component of the overall immune response and can facilitate pathogen clearance during a primary infection and also protect against subsequent infections. Dysregulation of the antibody response can lead to an autoimmune disease, malignancy, or enhanced infection. Since the experimental delineation of a distinct B cell lineage in 1965, various methods have been developed to understand antigen-specific B cell responses in the context of autoimmune diseases, primary immunodeficiencies, infection, and vaccination. In this review, we summarize the established techniques and discuss new and emerging technologies for probing the B cell response in vitro and in vivo by taking advantage of the specificity of B cell receptor (BCR)-associated and secreted antibodies. These include ELISPOT, flow cytometry, mass cytometry, and fluorescence microscopy to identify and/or isolate primary antigen-specific B cells. We also present our approach to identify rare antigen-specific B cells using magnetic enrichment followed by flow cytometry. Once these cells are isolated, in vitro proliferation assays and adoptive transfer experiments in mice can be used to further characterize antigen-specific B cell activation, function, and fate. Transgenic mouse models of B cells targeting model antigens and of B cell signaling have also significantly advanced our understanding of antigen-specific B cell responses in vivo.
Collapse
Affiliation(s)
- Jim Boonyaratanakornkit
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | | |
Collapse
|
20
|
Human Papillomavirus 16 Capsids Mediate Nuclear Entry during Infection. J Virol 2019; 93:JVI.00454-19. [PMID: 31092566 DOI: 10.1128/jvi.00454-19] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 05/04/2019] [Indexed: 12/27/2022] Open
Abstract
Infectious human papillomavirus 16 (HPV16) L1/L2 pseudovirions were found to remain largely intact during vesicular transport to the nucleus. By electron microscopy, capsids with a diameter of 50 nm were clearly visible within small vesicles attached to mitotic chromosomes and to a lesser extent within interphase nuclei, implying nuclear disassembly. By confocal analysis, it was determined that nuclear entry of assembled L1 is dependent upon the presence of the minor capsid protein, L2, but independent of encapsidated DNA. We also demonstrate that L1 nuclear localization and mitotic chromosome association can occur in vivo in the murine cervicovaginal challenge model of HPV16 infection. These findings challenge the prevailing concepts of PV uncoating and disassembly. More generally, they document that a largely intact viral capsid can enter the nucleus within a transport vesicle, establishing a novel mechanism by which a virus accesses the nuclear cellular machinery.IMPORTANCE Papillomaviruses (PVs) comprise a large family of nonenveloped DNA viruses that include HPV16, among other oncogenic types, the causative agents of cervical cancer. Delivery of the viral DNA into the host cell nucleus is necessary for establishment of infection. This was thought to occur via a subviral complex following uncoating of the larger viral capsid. In this study, we demonstrate that little disassembly of the PV capsid occurs prior to nuclear delivery. These surprising data reveal a previously unrecognized viral strategy to access the nuclear replication machinery. Understanding viral entry mechanisms not only increases our appreciation of basic cell biological pathways but also may lead to more effective antiviral interventions.
Collapse
|
21
|
Simonich CA, Doepker L, Ralph D, Williams JA, Dhar A, Yaffe Z, Gentles L, Small CT, Oliver B, Vigdorovich V, Mangala Prasad V, Nduati R, Sather DN, Lee KK, Matsen Iv FA, Overbaugh J. Kappa chain maturation helps drive rapid development of an infant HIV-1 broadly neutralizing antibody lineage. Nat Commun 2019; 10:2190. [PMID: 31097697 PMCID: PMC6522554 DOI: 10.1038/s41467-019-09481-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 03/08/2019] [Indexed: 12/14/2022] Open
Abstract
HIV-infected infants develop broadly neutralizing plasma responses with more rapid kinetics than adults, suggesting the ontogeny of infant responses could better inform a path to achievable vaccine targets. Here we reconstruct the developmental lineage of BF520.1, an infant-derived HIV-specific broadly neutralizing antibody (bnAb), using computational methods developed specifically for this purpose. We find that the BF520.1 inferred naive precursor binds HIV Env. We also show that heterologous cross-clade neutralizing activity evolved in the infant within six months of infection and that, ultimately, only 2% SHM is needed to achieve the full breadth of the mature antibody. Mutagenesis and structural analyses reveal that, for this infant bnAb, substitutions in the kappa chain were critical for activity, particularly in CDRL1. Overall, the developmental pathway of this infant antibody includes features distinct from adult antibodies, including several that may be amenable to better vaccine responses.
Collapse
Affiliation(s)
- Cassandra A Simonich
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
- Medical Scientist Training Program, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Laura Doepker
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Duncan Ralph
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - James A Williams
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, 98195, USA
| | - Amrit Dhar
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
- Department of Statistics, University of Washington, Seattle, WA, 98195, USA
| | - Zak Yaffe
- Medical Scientist Training Program, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Lauren Gentles
- Department of Microbiology, University of Washington, Seattle, WA, 98195, USA
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Christopher T Small
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Brian Oliver
- Center for Infectious Disease Research, Seattle, WA, 98109, USA
| | | | - Vidya Mangala Prasad
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, 98195, USA
| | - Ruth Nduati
- Department of Pediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - D Noah Sather
- Center for Infectious Disease Research, Seattle, WA, 98109, USA
| | - Kelly K Lee
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, 98195, USA
| | - Frederick A Matsen Iv
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Julie Overbaugh
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
- Medical Scientist Training Program, University of Washington School of Medicine, Seattle, WA, 98195, USA.
| |
Collapse
|
22
|
V H1-69 antiviral broadly neutralizing antibodies: genetics, structures, and relevance to rational vaccine design. Curr Opin Virol 2019; 34:149-159. [PMID: 30884330 DOI: 10.1016/j.coviro.2019.02.004] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 02/07/2019] [Indexed: 12/15/2022]
Abstract
Broadly neutralizing antibodies (bnAbs) are potential therapeutic molecules and valuable tools for studying conserved viral targets for vaccine and drug design. Interestingly, antibody responses to conserved epitopes can be highly convergent at the molecular level. Human antibodies targeting a number of viral antigens have often been found to utilize a restricted set of immunoglobulin germline genes in different individuals. Here we review recent knowledge on VH1-69-encoded antibodies in antiviral responses to influenza virus, HCV, and HIV-1. These antibodies share common genetic and structural features, and often develop neutralizing activity against a broad spectrum of viral strains. Understanding the genetic and structural characteristics of such antibodies and the target epitopes should help advance novel strategies to elicit bnAbs through vaccination.
Collapse
|
23
|
Longitudinal Analysis Reveals Early Development of Three MPER-Directed Neutralizing Antibody Lineages from an HIV-1-Infected Individual. Immunity 2019; 50:677-691.e13. [PMID: 30876875 DOI: 10.1016/j.immuni.2019.02.008] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 11/13/2018] [Accepted: 02/13/2019] [Indexed: 12/21/2022]
Abstract
Lineage-based vaccine design is an attractive approach for eliciting broadly neutralizing antibodies (bNAbs) against HIV-1. However, most bNAb lineages studied to date have features indicative of unusual recombination and/or development. From an individual in the prospective RV217 cohort, we identified three lineages of bNAbs targeting the membrane-proximal external region (MPER) of the HIV-1 envelope. Antibodies RV217-VRC42.01, -VRC43.01, and -VRC46.01 used distinct modes of recognition and neutralized 96%, 62%, and 30%, respectively, of a 208-strain virus panel. All three lineages had modest levels of somatic hypermutation and normal antibody-loop lengths and were initiated by the founder virus MPER. The broadest lineage, VRC42, was similar to the known bNAb 4E10. A multimeric immunogen based on the founder MPER activated B cells bearing the unmutated common ancestor of VRC42, with modest maturation of early VRC42 intermediates imparting neutralization breadth. These features suggest that VRC42 may be a promising template for lineage-based vaccine design.
Collapse
|
24
|
Bailey JR, Barnes E, Cox AL. Approaches, Progress, and Challenges to Hepatitis C Vaccine Development. Gastroenterology 2019; 156:418-430. [PMID: 30268785 PMCID: PMC6340767 DOI: 10.1053/j.gastro.2018.08.060] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 08/12/2018] [Accepted: 08/14/2018] [Indexed: 12/16/2022]
Abstract
Risk factors for hepatitis C virus (HCV) infection vary, and there were an estimated 1.75 million new cases worldwide in 2015. The World Health Organization aims for a 90% reduction in new HCV infections by 2030. An HCV vaccine would prevent transmission, regardless of risk factors, and significantly reduce the global burden of HCV-associated disease. Barriers to development include virus diversity, limited models for testing vaccines, and our incomplete understanding of protective immune responses. Although highly effective vaccines could prevent infection altogether, immune responses that increase the rate of HCV clearance and prevent chronic infection may be sufficient to reduce disease burden. Adjuvant envelope or core protein and virus-vectored nonstructural antigen vaccines have been tested in healthy volunteers who are not at risk for HCV infection; viral vectors encoding nonstructural proteins are the only vaccine strategy to be tested in at-risk individuals. Despite development challenges, a prophylactic vaccine is necessary for global control of HCV.
Collapse
Affiliation(s)
- Justin R. Bailey
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Eleanor Barnes
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine and the Oxford NIHR Biomedical Research Centre, Oxford University, UK
| | - Andrea L. Cox
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland,Reprint requests Address requests for reprints to: Andrea L. Cox, MD, PhD, Division of Infectious Diseases, Johns Hopkins University School of Medicine, 551 Rangos Building, 855 N Wolfe Street, Baltimore, Maryland 21205. fax: (443)769-1221.
| |
Collapse
|
25
|
Ellsworth GB, Lensing SY, Ogilvie CB, Lee JY, Goldstone SE, Berry-Lawhorn JM, Jay N, Stier EA, Logan JS, Einstein MH, Saah A, Mitsuyasu RT, Aboulafia D, Palefsky JM, Wilkin TJ. A delayed dose of quadrivalent human papillomavirus vaccine demonstrates immune memory in HIV-1-infected men. PAPILLOMAVIRUS RESEARCH (AMSTERDAM, NETHERLANDS) 2018; 6:11-14. [PMID: 29807211 PMCID: PMC6121157 DOI: 10.1016/j.pvr.2018.05.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 04/20/2018] [Accepted: 05/23/2018] [Indexed: 12/20/2022]
MESH Headings
- Adult
- Antibodies, Neutralizing/blood
- Antibodies, Viral/blood
- HIV Infections/complications
- HIV Infections/immunology
- Human Papillomavirus Recombinant Vaccine Quadrivalent, Types 6, 11, 16, 18/administration & dosage
- Human Papillomavirus Recombinant Vaccine Quadrivalent, Types 6, 11, 16, 18/immunology
- Humans
- Immunologic Memory
- Male
- Middle Aged
- Papillomavirus Infections/immunology
- Papillomavirus Infections/prevention & control
Collapse
Affiliation(s)
- G B Ellsworth
- Division of Infectious Diseases, Weill Cornell Medical College, 53W 23rd St, 6th Fl, New York, NY 10023, USA
| | - S Y Lensing
- University of Arkansas for Medical Sciences, Little Rock, USA
| | - C B Ogilvie
- Weill Cornell Medical College, New York City, USA
| | - J Y Lee
- University of Arkansas for Medical Sciences, Little Rock, USA
| | | | | | - N Jay
- University of California at San Francisco, USA
| | - E A Stier
- Boston University Medical Center, Boston, MA, USA
| | | | - M H Einstein
- Albert Einstein College of Medicine, USA; Albert Einstein Cancer Center, Montefiore Medical Center, New York City, USA
| | - A Saah
- Merck Laboratories, Whitehouse Station, NJ, USA
| | | | - D Aboulafia
- Virginia Mason Medical Center, Seattle, WA, USA; Division of Hematology, University of Washington, USA
| | | | - T J Wilkin
- Division of Infectious Diseases, Weill Cornell Medical College, 53W 23rd St, 6th Fl, New York, NY 10023, USA.
| |
Collapse
|
26
|
Schramm CA, Douek DC. Beyond Hot Spots: Biases in Antibody Somatic Hypermutation and Implications for Vaccine Design. Front Immunol 2018; 9:1876. [PMID: 30154794 PMCID: PMC6102386 DOI: 10.3389/fimmu.2018.01876] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/30/2018] [Indexed: 11/15/2022] Open
Abstract
The evolution of antibodies in an individual during an immune response by somatic hypermutation (SHM) is essential for the ability of the immune system to recognize and remove the diverse spectrum of antigens that may be encountered. These mutations are not produced at random; nucleotide motifs that result in increased or decreased rates of mutation were first reported in 1992. Newer models that estimate the propensity for mutation for every possible 5- or 7-nucleotide motif have emphasized the complexity of SHM targeting and suggested possible new hot spot motifs. Even with these fine-grained approaches, however, non-local context matters, and the mutations observed at a specific nucleotide motif varies between species and even by locus, gene segment, and position along the gene segment within a single species. An alternative method has been provided to further abstract away the molecular mechanisms underpinning SHM, prompted by evidence that certain stereotypical amino acid substitutions are favored at each position of a particular V gene. These "substitution profiles," whether obtained from a single B cell lineage or an entire repertoire, offer a simplified approach to predict which substitutions will be well-tolerated and which will be disfavored, without the need to consider path-dependent effects from neighboring positions. However, this comes at the cost of merging the effects of two distinct biological processes, the generation of mutations, and the selection acting on those mutations. Since selection is contingent on the particular antigens an individual has been exposed to, this suggests that SHM may have evolved to prefer mutations that are most likely to be useful against pathogens that have co-evolved with us. Alternatively, the ability to select favorable mutations may be strongly limited by the biases of SHM targeting. In either scenario, the sequence space explored by SHM is significantly limited and this consequently has profound implications for the rational design of vaccine strategies.
Collapse
Affiliation(s)
- Chaim A. Schramm
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, United States
| | - Daniel C. Douek
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, United States
| |
Collapse
|
27
|
Vectored gene delivery for lifetime animal contraception: Overview and hurdles to implementation. Theriogenology 2018; 112:63-74. [DOI: 10.1016/j.theriogenology.2017.11.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 10/25/2017] [Accepted: 11/02/2017] [Indexed: 12/24/2022]
|
28
|
Weinberg A, Huang S, Moscicki AB, Saah A, Levin MJ. Persistence of memory B-cell and T-cell responses to the quadrivalent HPV vaccine in HIV-infected children. AIDS 2018; 32:851-860. [PMID: 29424778 PMCID: PMC5869173 DOI: 10.1097/qad.0000000000001773] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To determine the magnitude and persistence of quadrivalent human papillomavirus (HPV)16 and HPV18 B-cell and T-cell memory after three or four doses of quadrivalent HPV vaccine (QHPV) in HIV-infected children. METHODS Seventy-four HIV-infected children immunized with four doses and 23 with three doses of QHPV had HPV16 and HPV18 IgG B-cell and IFNγ and IL2 T-cell ELISPOT performed at 2, 3.5 and 4-5 years after the last dose. RESULTS HPV16 and HPV18 T-cell responses were similar in both treatment groups, with higher responses to HPV16 vs. HPV18. These HPV T-cell responses correlated with HIV disease characteristics at the study visits. Global T-cell function declined over time as measured by nonspecific mitogenic stimulation. B-cell memory was similar across treatment groups and HPV genotypes. There was a decline in HPV-specific B-cell memory over time that reached statistical significance for HPV16 in the four-dose group. CONCLUSION B-cell and T-cell memory did not significantly differ after either three or four doses of QHPV in HIV-infected children. The clinical consequences of decreasing global T-cell function and HPV B-cell memory over time in HIV-infected children requires further investigation.
Collapse
Affiliation(s)
- Adriana Weinberg
- Section of Pediatric Infectious Diseases, Departments of Pediatrics and Medicine, and Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Sharon Huang
- Center for Biostatistics in AIDS Research, Harvard School of Public Health, Boston, Massachusetts
| | - Anna-Barbara Moscicki
- Department of Pediatrics, University of California Los Angeles, Los Angeles, California
| | | | - Myron J Levin
- Section of Pediatric Infectious Diseases, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
29
|
Stanley M. Tumour virus vaccines: hepatitis B virus and human papillomavirus. Philos Trans R Soc Lond B Biol Sci 2018; 372:rstb.2016.0268. [PMID: 28893935 DOI: 10.1098/rstb.2016.0268] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2017] [Indexed: 12/11/2022] Open
Abstract
Two of the most important human oncogenic viruses are hepatitis B virus (HBV) and human papillomavirus (HPV). HBV infection has been preventable by vaccination since 1982; vaccination of neonates and infants is highly effective, resulting already in decreased rates of new infections, chronic liver disease and hepato-cellular carcinoma. Nonetheless, HBV remains a global public health problem with high rates of vertical transmission from mother to child in some regions. Prophylactic HPV vaccines composed of virus-like particles (VLPs) of the L1 capsid protein have been licensed since 2006/2007. These target infection by the oncogenic HPVs 16 and 18 (the cause of 70% of cervical cancers); a new vaccine licensed in 2014/2015 additionally targets HPVs 31, 33, 45, 52, 58. HPV vaccines are now included in the national immunization programmes in many countries, with young adolescent peri-pubertal girls the usual cohort for immunization. Population effectiveness in women is now being demonstrated in countries with high vaccine coverage with significant reductions in high-grade cervical intra-epithelial neoplasia (a surrogate for cervical cancer), genital warts and vaccine HPV type genoprevalence. Herd effects in young heterosexual men and older women are evident. Cancers caused by HBV and HPV should, in theory, be amenable to immunotherapies and various therapeutic vaccines for HPV in particular are in development and/or in clinical trial.This article is part of the themed issue 'Human oncogenic viruses'.
Collapse
Affiliation(s)
- Margaret Stanley
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| |
Collapse
|
30
|
Abstract
The discovery of genotype 16 as the prototype oncogenic human papillomavirus (HPV) initiated a quarter century of laboratory and epidemiological studies that demonstrated their necessary, but not sufficient, aetiological role in cervical and several other anogenital and oropharyngeal cancers. Early virus-induced immune deviation can lead to persistent subclinical infection that brings the risk of progression to cancer. Effective secondary prevention of cervical cancer through cytological and/or HPV screening depends on regular and widespread use in the general population, but coverage is inadequate in low-resource settings. The discovery that the major capsid antigen L1 could self-assemble into empty virus-like particles (VLPs) that are both highly immunogenic and protective led to the licensure of several prophylactic VLP-based HPV vaccines for the prevention of cervical cancer. The implementation of vaccination programmes in adolescent females is underway in many countries, but their impact critically depends on the population coverage and is improved by herd immunity. This Review considers how our expanding knowledge of the virology and immunology of HPV infection can be exploited to improve vaccine technologies and delivery of such preventive strategies to maximize reductions in HPV-associated disease, including incorporation of an HPV vaccine covering oncogenic types within a standard multitarget paediatric vaccine.
Collapse
Affiliation(s)
| | - Peter L. Stern
- Division of Molecular and Clinical Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
31
|
Scherer EM, Smith RA, Carter JJ, Wipf GC, Gallego DF, Stern M, Wald A, Galloway DA. Analysis of Memory B-Cell Responses Reveals Suboptimal Dosing Schedule of a Licensed Vaccine. J Infect Dis 2018; 217:572-580. [PMID: 29186468 PMCID: PMC5853470 DOI: 10.1093/infdis/jix566] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Current guidance recommends that adolescents receive a 2-dose human papillomavirus (HPV) vaccine, whereas young adults and immunocompromised persons receive 3 doses. We examined secondary responses of vaccine-elicited memory B cells (Bmem) in naive women receiving 3 doses of the quadrivalent HPV vaccine to understand the quality of B-cell memory generated by this highly effective vaccine. Unexpectedly, we observed a lower Bmem response rate and magnitude of Bmem responses to the third dose than to a booster dose administered at month 24. Moreover, high titers of antigen-specific serum antibody at vaccination inversely correlated with Bmem responses. As the purpose of additional doses/boosters is to stimulate Bmem to rapidly boost antibody levels, these results indicate the timing of the third dose is suboptimal and lend support to a 2-dose HPV vaccine for young adults. Our findings also indicate more broadly that multidose vaccine schedules should be rationally determined on the basis of Bmem responses.
Collapse
Affiliation(s)
- Erin M Scherer
- Human Biology Division, Fred Hutchinson Cancer Research Center Seattle, Washington
| | - Robin A Smith
- Human Biology Division, Fred Hutchinson Cancer Research Center Seattle, Washington
| | - Joseph J Carter
- Human Biology Division, Fred Hutchinson Cancer Research Center Seattle, Washington
| | - Gregory C Wipf
- Human Biology Division, Fred Hutchinson Cancer Research Center Seattle, Washington
| | - Daniel F Gallego
- Human Biology Division, Fred Hutchinson Cancer Research Center Seattle, Washington
| | | | - Anna Wald
- Department of Medicine, Seattle, Washington
- Department of Laboratory Medicine, Seattle, Washington
- Department of Epidemiology, University of Washington; Seattle, Washington
- Department of Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center; Seattle, Washington
| | - Denise A Galloway
- Human Biology Division, Fred Hutchinson Cancer Research Center Seattle, Washington
- Department of Microbiology, University of Washington, Seattle, Washington
| |
Collapse
|
32
|
Abstract
Antibodies have been used for over a century prophylactically and, less often, therapeutically against viruses. 'Super-antibodies' — a new generation of highly potent and/or broadly cross-reactive human monoclonal antibodies — offer new opportunities for prophylaxis and therapy of viral infections. Super-antibodies are typically generated infrequently and/or in a limited number of individuals during natural infections. Isolation of these antibodies has primarily been achieved by large-scale screening for suitable donors and new single B cell approaches to human monoclonal antibody generation. Super-antibodies may offer the possibility of treating multiple viruses of a given family with a single reagent. They are also valuable templates for rational vaccine design. The great potency of super-antibodies has many advantages for practical development as therapeutic reagents. These advantages can be enhanced by a variety of antibody engineering technologies.
So-called super-antibodies are highly potent, broadly reactive antiviral antibodies that offer promise for the treatment of various chronic and emerging viruses. This Review describes how recent technological advances led to their isolation from rare, infected individuals and their development for the prevention and treatment of various viral infections. Antibodies have been used for more than 100 years in the therapy of infectious diseases, but a new generation of highly potent and/or broadly cross-reactive human monoclonal antibodies (sometimes referred to as 'super-antibodies') offers new opportunities for intervention. The isolation of these antibodies, most of which are rarely induced in human infections, has primarily been achieved by large-scale screening for suitable donors and new single B cell approaches to human monoclonal antibody generation. Engineering the antibodies to improve half-life and effector functions has further augmented their in vivo activity in some cases. Super-antibodies offer promise for the prophylaxis and therapy of infections with a range of viruses, including those that are highly antigenically variable and those that are newly emerging or that have pandemic potential. The next few years will be decisive in the realization of the promise of super-antibodies.
Collapse
|
33
|
Antibody persistence and evidence of immune memory at 5 years following administration of the 9-valent HPV vaccine. Vaccine 2017; 35:5050-5057. [DOI: 10.1016/j.vaccine.2017.07.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 07/03/2017] [Accepted: 07/05/2017] [Indexed: 12/21/2022]
|
34
|
Karlsson Hedestam GB, Guenaga J, Corcoran M, Wyatt RT. Evolution of B cell analysis and Env trimer redesign. Immunol Rev 2017; 275:183-202. [PMID: 28133805 DOI: 10.1111/imr.12515] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
HIV-1 and its surface envelope glycoproteins (Env), gp120 and gp41, have evolved immune evasion strategies that render the elicitation of effective antibody responses to the functional Env entry unit extremely difficult. HIV-1 establishes chronic infection and stimulates vigorous immune responses in the human host; forcing selection of viral variants that escape cellular and antibody (Ab)-mediated immune pressure, yet possess contemporary fitness. Successful survival of fit variants through the gauntlet of the human immune system make this virus and these glycoproteins a formidable challenge to target by vaccination, requiring a systematic approach to Env mimetic immunogen design and evaluation of elicited responses. Here, we review key aspects of HIV-1 Env immunogenicity and immunogen re-design, based on experimental data generated by us and others over the past decade or more. We further provide rationale and details regarding the use of newly evolving tools to analyze B cell responses, including approaches to use next generation sequencing for antibody lineage tracing and B cell fate mapping. Together, these developments offer opportunities to address long-standing questions about the establishment of effective B cell immunity elicited by vaccination, not just against HIV-1.
Collapse
Affiliation(s)
| | - Javier Guenaga
- Department of Immunology and Microbial Science, IAVI Neutralizing Antibody Center at TSRI, La Jolla, CA, USA
| | - Martin Corcoran
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Richard T Wyatt
- Department of Immunology and Microbial Science, IAVI Neutralizing Antibody Center at TSRI, La Jolla, CA, USA.,The Scripps CHAVI-ID, La Jolla, CA, USA
| |
Collapse
|
35
|
Antibody therapies for the prevention and treatment of viral infections. NPJ Vaccines 2017; 2:19. [PMID: 29263875 PMCID: PMC5627241 DOI: 10.1038/s41541-017-0019-3] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 05/08/2017] [Accepted: 05/16/2017] [Indexed: 12/18/2022] Open
Abstract
Antibodies are an important component in host immune responses to viral pathogens. Because of their unique maturation process, antibodies can evolve to be highly specific to viral antigens. Physicians and researchers have been relying on such high specificity in their quest to understand host–viral interaction and viral pathogenesis mechanisms and to find potential cures for viral infection and disease. With more than 60 recombinant monoclonal antibodies developed for human use in the last 20 years, monoclonal antibodies are now considered a viable therapeutic modality for infectious disease targets, including newly emerging viral pathogens such as Ebola representing heightened public health concerns, as well as pathogens that have long been known, such as human cytomegalovirus. Here, we summarize some recent advances in identification and characterization of monoclonal antibodies suitable as drug candidates for clinical evaluation, and review some promising candidates in the development pipeline.
Collapse
|
36
|
Kenney M, Waters RA, Rieder E, Pega J, Perez-Filguera M, Golde WT. Enhanced sensitivity in detection of antiviral antibody responses using biotinylation of foot-and-mouth disease virus (FMDV) capsids. J Immunol Methods 2017; 450:1-9. [PMID: 28689695 DOI: 10.1016/j.jim.2017.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 06/23/2017] [Accepted: 07/05/2017] [Indexed: 01/28/2023]
Abstract
Analysis of the immune response to infection of livestock by foot-and-mouth disease virus (FMDV) is most often reported as the serum antibody response to the virus. While measurement of neutralizing antibody has been sensitive and specific, measurements of the quality of the antibody response are less robust. Determining the immunoglobulin (Ig) isotype of the serum antibody response provides a deeper understanding of the biology of the response and more sensitive methods for these assays will facilitate analyses of B cell mediated immunity. We tested the hypothesis that using the virus as the molecular probe could be achieved by adding tags to the surface of the FMDV capsid, and that would enhance sensitivity in assays for anti-FMDV antibody responses. The use of a FLAG-tagged virus in these assays failed to yield improvement whereas chemically biotinylating the virus capsid resulted in significant enhancement of the signal. Here we describe methods using biotinylated virus for measuring anti-viral antibody in serum and antibody secreting cells (ASCs) in blood that are sensitive and specific. Finally, we describe using the biotinylated virus in flow cytometry where such assays should greatly enhance the analysis of anti-virus antibody producing B cells, allowing the investigator to focus on only the FMDV specific B cells when analyzing the development of the B cell response to either infection or vaccination.
Collapse
Affiliation(s)
- Mary Kenney
- Plum Island Animal Disease Center, Agricultural Research Service, USDA, Greenport, NY 11944, United States
| | - Ryan A Waters
- Plum Island Animal Disease Center, Agricultural Research Service, USDA, Greenport, NY 11944, United States
| | - Elizabeth Rieder
- Plum Island Animal Disease Center, Agricultural Research Service, USDA, Greenport, NY 11944, United States
| | - Juan Pega
- Instituto de Virología, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas (CICVyA), Instituto Nacional de Tecnología Agropecuaria (INTA), Buenos Aires, Argentina
| | - Mariano Perez-Filguera
- Instituto de Virología, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas (CICVyA), Instituto Nacional de Tecnología Agropecuaria (INTA), Buenos Aires, Argentina
| | - William T Golde
- Plum Island Animal Disease Center, Agricultural Research Service, USDA, Greenport, NY 11944, United States.
| |
Collapse
|
37
|
Bailey JR, Flyak AI, Cohen VJ, Li H, Wasilewski LN, Snider AE, Wang S, Learn GH, Kose N, Loerinc L, Lampley R, Cox AL, Pfaff JM, Doranz BJ, Shaw GM, Ray SC, Crowe JE. Broadly neutralizing antibodies with few somatic mutations and hepatitis C virus clearance. JCI Insight 2017; 2:92872. [PMID: 28469084 PMCID: PMC5414559 DOI: 10.1172/jci.insight.92872] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 03/21/2017] [Indexed: 01/15/2023] Open
Abstract
Here, we report the isolation of broadly neutralizing mAbs (bNAbs) from persons with broadly neutralizing serum who spontaneously cleared hepatitis C virus (HCV) infection. We found that bNAbs from two donors bound the same epitope and were encoded by the same germline heavy chain variable gene segment. Remarkably, these bNAbs were encoded by antibody variable genes with sparse somatic mutations. For one of the most potent bNAbs, these somatic mutations were critical for antibody neutralizing breadth and for binding to autologous envelope variants circulating late in infection. However, somatic mutations were not necessary for binding of the bNAb unmutated ancestor to envelope proteins of early autologous transmitted/founder viruses. This study identifies a public B cell clonotype favoring early recognition of a conserved HCV epitope, proving that anti-HCV bNAbs can achieve substantial neutralizing breadth with relatively few somatic mutations, and identifies HCV envelope variants that favored selection and maturation of an anti-HCV bNAb in vivo. These data provide insight into the molecular mechanisms of immune-mediated clearance of HCV infection and present a roadmap to guide development of a vaccine capable of stimulating anti-HCV bNAbs with a physiologic number of somatic mutations characteristic of vaccine responses.
Collapse
Affiliation(s)
- Justin R. Bailey
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Andrew I. Flyak
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Valerie J. Cohen
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hui Li
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lisa N. Wasilewski
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Anna E. Snider
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Shuyi Wang
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gerald H. Learn
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nurgun Kose
- Vanderbilt Vaccine Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Leah Loerinc
- Vanderbilt Vaccine Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Rebecca Lampley
- Vanderbilt Vaccine Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Andrea L. Cox
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland, USA
| | | | | | - George M. Shaw
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Stuart C. Ray
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland, USA
| | - James E. Crowe
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Vaccine Center, Vanderbilt University, Nashville, Tennessee, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
38
|
Abstract
The induction of neutralizing antibodies directed against the human immunodeficiency virus (HIV) has received considerable attention in recent years, in part driven by renewed interest and opportunities for antibody-based strategies for prevention such as passive transfer of antibodies and the development of preventive vaccines, as well as immune-based therapeutic interventions. Advances in the ability to screen, isolate, and characterize HIV-specific antibodies have led to the identification of a new generation of potent broadly neutralizing antibodies (bNAbs). The majority of these antibodies have been isolated from B cells of chronically HIV-infected individuals with detectable viremia. In this review, we provide insight into the phenotypic and functional attributes of human B cells, with a focus on HIV-specific memory B cells and plasmablasts/cells that are responsible for sustaining humoral immune responses against HIV. We discuss the abnormalities in B cells that occur in HIV infection both in the peripheral blood and lymphoid tissues, especially in the setting of persisting viremia. Finally, we consider the opportunities and drawbacks of intensively interrogating antibodies isolated from HIV-infected individuals to guide strategies aimed at developing effective antibody-based vaccine and therapeutic interventions for HIV.
Collapse
Affiliation(s)
- Susan Moir
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892
| | - Anthony S. Fauci
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892
| |
Collapse
|
39
|
Deconstructing the Antiviral Neutralizing-Antibody Response: Implications for Vaccine Development and Immunity. Microbiol Mol Biol Rev 2016; 80:989-1010. [PMID: 27784796 DOI: 10.1128/mmbr.00024-15] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The antibody response plays a key role in protection against viral infections. While antiviral antibodies may reduce the viral burden via several mechanisms, the ability to directly inhibit (neutralize) infection of cells has been extensively studied. Eliciting a neutralizing-antibody response is a goal of many vaccine development programs and commonly correlates with protection from disease. Considerable insights into the mechanisms of neutralization have been gained from studies of monoclonal antibodies, yet the individual contributions and dynamics of the repertoire of circulating antibody specificities elicited by infection and vaccination are poorly understood on the functional and molecular levels. Neutralizing antibodies with the most protective functionalities may be a rare component of a polyclonal, pathogen-specific antibody response, further complicating efforts to identify the elements of a protective immune response. This review discusses advances in deconstructing polyclonal antibody responses to flavivirus infection or vaccination. Our discussions draw comparisons to HIV-1, a virus with a distinct structure and replication cycle for which the antibody response has been extensively investigated. Progress toward deconstructing and understanding the components of polyclonal antibody responses identifies new targets and challenges for vaccination strategies.
Collapse
|
40
|
Minimally Mutated HIV-1 Broadly Neutralizing Antibodies to Guide Reductionist Vaccine Design. PLoS Pathog 2016; 12:e1005815. [PMID: 27560183 PMCID: PMC4999182 DOI: 10.1371/journal.ppat.1005815] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 07/19/2016] [Indexed: 11/19/2022] Open
Abstract
An optimal HIV vaccine should induce broadly neutralizing antibodies (bnAbs) that neutralize diverse viral strains and subtypes. However, potent bnAbs develop in only a small fraction of HIV-infected individuals, all contain rare features such as extensive mutation, insertions, deletions, and/or long complementarity-determining regions, and some are polyreactive, casting doubt on whether bnAbs to HIV can be reliably induced by vaccination. We engineered two potent VRC01-class bnAbs that minimized rare features. According to a quantitative features frequency analysis, the set of features for one of these minimally mutated bnAbs compared favorably with all 68 HIV bnAbs analyzed and was similar to antibodies elicited by common vaccines. This same minimally mutated bnAb lacked polyreactivity in four different assays. We then divided the minimal mutations into spatial clusters and dissected the epitope components interacting with those clusters, by mutational and crystallographic analyses coupled with neutralization assays. Finally, by synthesizing available data, we developed a working-concept boosting strategy to select the mutation clusters in a logical order following a germline-targeting prime. We have thus developed potent HIV bnAbs that may be more tractable vaccine goals compared to existing bnAbs, and we have proposed a strategy to elicit them. This reductionist approach to vaccine design, guided by antibody and antigen structure, could be applied to design candidate vaccines for other HIV bnAbs or protective Abs against other pathogens. Many HIV vaccine design efforts aim to elicit so-called broadly neutralizing antibodies that bind and neutralize diverse strains and subtypes of the virus. However, these efforts are guided by very unusual antibodies isolated from HIV-infected individuals. These antibodies have rare features that limit their use as direct vaccine templates, because it is unlikely that any vaccine could consistently elicit similar antibodies. We engineered HIV broadly neutralizing antibodies that minimized these rare features and may therefore serve as better leads for HIV vaccine design. Antibodies generally gain affinity for their target epitope by accumulating mutations in a natural process of maturation. Figuring out how to use vaccines to elicit particular kinds of antibodies, with particular kinds of helpful mutations, is a major unsolved challenge for vaccine design. We were able to determine which mutations in our new antibodies are most important and which epitope structures are needed to induce those mutations. This analysis allowed us to deduce a logical strategy, which remains to be tested, for how to guide the maturation of these types of antibodies by vaccination. We propose that this reductionist approach to vaccine design, guided by molecular structure and engineering-oriented to allow for optimization, has promise for designing vaccines against HIV and many other pathogens.
Collapse
|
41
|
Affiliation(s)
- John T Schiller
- Center for Cancer Research, National Cancer Institute, NIH, United States.
| |
Collapse
|
42
|
Seifert M, Küppers R. Human memory B cells. Leukemia 2016; 30:2283-2292. [DOI: 10.1038/leu.2016.226] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 06/29/2016] [Accepted: 07/26/2016] [Indexed: 12/20/2022]
|
43
|
Scherer EM, Smith RA, Gallego DF, Carter JJ, Wipf GC, Hoyos M, Stern M, Thurston T, Trinklein ND, Wald A, Galloway DA. A Single Human Papillomavirus Vaccine Dose Improves B Cell Memory in Previously Infected Subjects. EBioMedicine 2016; 10:55-64. [PMID: 27423190 PMCID: PMC5006801 DOI: 10.1016/j.ebiom.2016.06.042] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 06/24/2016] [Accepted: 06/27/2016] [Indexed: 02/06/2023] Open
Abstract
Although licensed human papillomavirus (HPV) vaccines are most efficacious in persons never infected with HPV, they also reduce infection and disease in previously infected subjects, indicating natural immunity is not entirely protective against HPV re-infection. The aim of this exploratory study was to examine the B cell memory elicited by HPV infection and evaluate whether vaccination merely boosts antibody (Ab) levels in previously infected subjects or also improves the quality of B cell memory. Toward this end, the memory B cells (Bmem) of five unvaccinated, HPV-seropositive subjects were isolated and characterized, and subject recall responses to a single HPV vaccine dose were analyzed. Vaccination boosted Ab levels 24- to 930-fold (median 77-fold) and Bmem numbers 3- to 27-fold (median 6-fold). In addition, Abs cloned from naturally elicited Bmem were generally non-neutralizing, whereas all those isolated following vaccination were neutralizing. Moreover, Ab and plasmablast responses indicative of memory recall responses were only observed in two subjects. These results suggest HPV vaccination augments both the magnitude and quality of natural immunity and demonstrate that sexually active persons could also benefit from HPV vaccination. This study may have important public policy implications, especially for the older 'catch-up' group within the vaccine's target population.
Collapse
Affiliation(s)
- Erin M Scherer
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Robin A Smith
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Daniel F Gallego
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Joseph J Carter
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Gregory C Wipf
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Manuela Hoyos
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Michael Stern
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Tate Thurston
- Panoply Bio, a division of Active Motif, Carlsbad, CA 92008, USA
| | | | - Anna Wald
- Department of Medicine, University of Washington, Seattle, WA 98195, USA; Department of Laboratory Medicine, University of Washington, Seattle, WA 98195, USA; Department of Epidemiology, University of Washington, Seattle, WA 98195, USA; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Denise A Galloway
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Microbiology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
44
|
Simonich CA, Williams KL, Verkerke HP, Williams JA, Nduati R, Lee KK, Overbaugh J. HIV-1 Neutralizing Antibodies with Limited Hypermutation from an Infant. Cell 2016; 166:77-87. [PMID: 27345369 DOI: 10.1016/j.cell.2016.05.055] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 04/19/2016] [Accepted: 05/17/2016] [Indexed: 12/23/2022]
Abstract
HIV-1 broadly neutralizing antibodies (bnAbs) develop in a subset of infected adults and exhibit high levels of somatic hypermutation (SHM) due to years of affinity maturation. There is no precedent for eliciting highly mutated antibodies by vaccination, nor is it practical to wait years for a desired response. Infants develop broad responses early, which may suggest a more direct path to generating bnAbs. Here, we isolated ten neutralizing antibodies (nAbs) contributing to plasma breadth of an infant at ∼1 year post-infection, including one with cross-clade breadth. The nAbs bind to envelope trimer from the transmitted virus, suggesting that this interaction may have initiated development of the infant nAbs. The infant cross-clade bnAb targets the N332 supersite on envelope but, unlike adult bnAbs targeting this site, lacks indels and has low SHM. The identification of this infant bnAb illustrates that HIV-1-specific neutralization breadth can develop without prolonged affinity maturation and extensive SHM.
Collapse
Affiliation(s)
- Cassandra A Simonich
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Medical Scientist Training Program, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Katherine L Williams
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Hans P Verkerke
- Department of Medicinal Chemistry, University of Washington, Seattle, WA 98195, USA
| | - James A Williams
- Department of Medicinal Chemistry, University of Washington, Seattle, WA 98195, USA
| | - Ruth Nduati
- Department of Pediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Kelly K Lee
- Department of Medicinal Chemistry, University of Washington, Seattle, WA 98195, USA
| | - Julie Overbaugh
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Medical Scientist Training Program, University of Washington School of Medicine, Seattle, WA 98195, USA.
| |
Collapse
|
45
|
Pejoski D, Tchitchek N, Rodriguez Pozo A, Elhmouzi-Younes J, Yousfi-Bogniaho R, Rogez-Kreuz C, Clayette P, Dereuddre-Bosquet N, Lévy Y, Cosma A, Le Grand R, Beignon AS. Identification of Vaccine-Altered Circulating B Cell Phenotypes Using Mass Cytometry and a Two-Step Clustering Analysis. THE JOURNAL OF IMMUNOLOGY 2016; 196:4814-31. [DOI: 10.4049/jimmunol.1502005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 03/31/2016] [Indexed: 01/08/2023]
|
46
|
Stanley M. HPV vaccines: how many doses are needed for protection? Future Virol 2016. [DOI: 10.2217/fvl-2016-0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
HPV virus-like particle vaccines are highly immunogenic, well tolerated and are in the national immunization programs in more than 62 countries. Genital HPV is a sexually transmitted infection with first infection occurring just after the onset of sexual activity. The routine cohort for immunization in almost all countries are adolescent girls 9–15 years of age with or without catch-up for older adolescents and young women. In countries with vaccine coverage exceeding 50%, reductions in vaccine type HPV geno-prevalence and disease are being shown. The mechanism of protection is assumed to be via neutralizing antibody. Antibody concentration in adolescents less than 14 years of age after two doses of vaccine at 0 and 6 months are noninferior to women after three doses and in whom efficacy was demonstrated in randomized control trials. The original three-dose schedules have already been reduced in many countries, for those 14 years of age and under, to two doses at least 6 months apart for the licensed vaccines Cervarix® and GARDASIL®. There is preliminary evidence that one dose of vaccine is as effective as two or three doses at preventing persistent HPV infection in the cervix in young women and a one-dose schedule may be possible if supported by evidence from randomized controlled trials.
Collapse
Affiliation(s)
- Margaret Stanley
- Department of Pathology, Tennis Court Road, Cambridge, CB2 1QP, UK
| |
Collapse
|
47
|
Xia L, Xian Y, Wang D, Chen Y, Huang X, Bi X, Yu H, Fu Z, Liu X, Li S, An Z, Luo W, Zhao Q, Xia N. A human monoclonal antibody against HPV16 recognizes an immunodominant and neutralizing epitope partially overlapping with that of H16.V5. Sci Rep 2016; 6:19042. [PMID: 26750243 PMCID: PMC4707464 DOI: 10.1038/srep19042] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 12/03/2015] [Indexed: 11/13/2022] Open
Abstract
The presence of neutralizing epitopes in human papillomavirus (HPV) L1 virus-like particles (VLPs) is the structural basis of prophylactic vaccines. An anti-HPV16 neutralizing monoclonal antibody (N-mAb) 26D1 was isolated from a memory B cell of a human vaccinee. The pre-binding of heparan sulfate to VLPs inhibited the binding of both N-mAbs to the antigen, indicating that the epitopes are critical for viral cell attachment/entry. Hybrid VLP binding with surface loop swapping between types indicated the essential roles of the DE and FG loops for both 26D1 (DEa in particular) and H16.V5 binding. Specifically, Tyr(135) and Val(141) on the DEa loop were shown to be critical residues for 26D1 binding via site-directed mutagenesis. Partially overlap between the epitopes between 26D1 and H16.V5 was shown using pairwise epitope mapping, and their binding difference is demonstrated to be predominantly in DE loop region. In addition, 26D1 epitope is immunodominant epitope recognized by both antibodies elicited by the authentic virus from infected individuals and polyclonal antibodies from vaccinees. Overall, a partially overlapping but distinct neutralizing epitope from that of H16.V5 was identified using a human N-mAb, shedding lights to the antibody arrays as part of human immune response to vaccination and infection.
Collapse
Affiliation(s)
- Lin Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Science, Xiamen University; Xiamen 361105, China
| | - Yangfei Xian
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Science, Xiamen University; Xiamen 361105, China
| | - Daning Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Science, Xiamen University; Xiamen 361105, China
| | - Yuanzhi Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Science, Xiamen University; Xiamen 361105, China
| | - Xiaofen Huang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Science, Xiamen University; Xiamen 361105, China
| | - Xingjian Bi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Science, Xiamen University; Xiamen 361105, China
| | - Hai Yu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Science, Xiamen University; Xiamen 361105, China
| | - Zheng Fu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Science, Xiamen University; Xiamen 361105, China
| | - Xinlin Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Science, Xiamen University; Xiamen 361105, China
| | - Shaowei Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Science, Xiamen University; Xiamen 361105, China
| | - Zhiqiang An
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Science, Xiamen University; Xiamen 361105, China
- Texas Therapeutics Institute, The Brown Foundation of Molecular Medicine, University of Texas Health Science Center at Houston, Houston TX77030, USA
| | - Wenxin Luo
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Science, Xiamen University; Xiamen 361105, China
| | - Qinjian Zhao
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Science, Xiamen University; Xiamen 361105, China
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Science, Xiamen University; Xiamen 361105, China
| |
Collapse
|
48
|
Cox KS, Tang A, Chen Z, Horton MS, Yan H, Wang XM, Dubey SA, DiStefano DJ, Ettenger A, Fong RH, Doranz BJ, Casimiro DR, Vora KA. Rapid isolation of dengue-neutralizing antibodies from single cell-sorted human antigen-specific memory B-cell cultures. MAbs 2015; 8:129-40. [PMID: 26491897 PMCID: PMC4966506 DOI: 10.1080/19420862.2015.1109757] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Monitoring antigen-specific memory B cells and the antibodies they encode is important for understanding the specificity, breadth and duration of immune response to an infection or vaccination. The antibodies isolated could further help design vaccine antigens for raising relevant protective immune responses. However, developing assays to measure and isolate antigen-specific memory B cells is technically challenging due to the low frequencies of these cells that exist in the circulating blood. Here, we describe a flow cytometry method to identify and isolate dengue envelope-specific memory B cells using a labeled dengue envelope protein. We enumerated dengue-envelope specific memory B cells from a cohort of dengue seropositive donors using this direct flow cytometry assay. A more established and conventional assay, the cultured B ELISPOT, was used as a benchmark comparator. Furthermore, we were able to confirm the single-sorted memory B-cell specificity by culturing B cells and differentiating them into plasma cells using cell lines expressing CD40L. The culture supernatants were assayed for antigen binding and the ability of the antibodies to neutralize the cognate dengue virus. Moreover, we successfully isolated the heavy and light Ig sequences and expressed them as full-length recombinant antibodies to reproduce the activity seen in culture supernatants. Mapping of these antibodies revealed a novel epitope for dengue 2 virus serotype. In conclusion, we established a reproducible methodology to enumerate antigen-specific memory B cells and assay their encoded antibodies for functional characterization.
Collapse
Affiliation(s)
- Kara S Cox
- a Department of Infectious Diseases and Vaccines-West Point ; PA; Merck Research Laboratories; Merck & Co.; Inc. Kenilworth ; NJ ; USA
| | - Aimin Tang
- a Department of Infectious Diseases and Vaccines-West Point ; PA; Merck Research Laboratories; Merck & Co.; Inc. Kenilworth ; NJ ; USA
| | - Zhifeng Chen
- a Department of Infectious Diseases and Vaccines-West Point ; PA; Merck Research Laboratories; Merck & Co.; Inc. Kenilworth ; NJ ; USA
| | - Melanie S Horton
- a Department of Infectious Diseases and Vaccines-West Point ; PA; Merck Research Laboratories; Merck & Co.; Inc. Kenilworth ; NJ ; USA
| | - Hao Yan
- a Department of Infectious Diseases and Vaccines-West Point ; PA; Merck Research Laboratories; Merck & Co.; Inc. Kenilworth ; NJ ; USA
| | - Xin-Min Wang
- a Department of Infectious Diseases and Vaccines-West Point ; PA; Merck Research Laboratories; Merck & Co.; Inc. Kenilworth ; NJ ; USA
| | - Sheri A Dubey
- a Department of Infectious Diseases and Vaccines-West Point ; PA; Merck Research Laboratories; Merck & Co.; Inc. Kenilworth ; NJ ; USA
| | - Daniel J DiStefano
- a Department of Infectious Diseases and Vaccines-West Point ; PA; Merck Research Laboratories; Merck & Co.; Inc. Kenilworth ; NJ ; USA
| | | | | | | | - Danilo R Casimiro
- a Department of Infectious Diseases and Vaccines-West Point ; PA; Merck Research Laboratories; Merck & Co.; Inc. Kenilworth ; NJ ; USA
| | - Kalpit A Vora
- a Department of Infectious Diseases and Vaccines-West Point ; PA; Merck Research Laboratories; Merck & Co.; Inc. Kenilworth ; NJ ; USA
| |
Collapse
|