Immune checkpoint modulation enhances HIV-1 antibody induction.

Abstract

Eliciting protective titers of HIV-1 broadly neutralizing antibodies (bnAbs) is a goal of HIV-1 vaccine development, but current vaccine strategies have yet to induce bnAbs in humans. Many bnAbs isolated from HIV-1-infected individuals are encoded by immunoglobulin gene rearrangments with infrequent naive B cell precursors and with unusual genetic features that may be subject to host regulatory control. Here, we administer antibodies targeting immune cell regulatory receptors CTLA-4, PD-1 or OX40 along with HIV envelope (Env) vaccines to rhesus macaques and bnAb immunoglobulin knock-in (KI) mice expressing diverse precursors of CD4 binding site HIV-1 bnAbs. CTLA-4 blockade augments HIV-1 Env antibody responses in macaques, and in a bnAb-precursor mouse model, CTLA-4 blocking or OX40 agonist antibodies increase germinal center B and T follicular helper cells and plasma neutralizing antibodies. Thus, modulation of CTLA-4 or OX40 immune checkpoints during vaccination can promote germinal center activity and enhance HIV-1 Env antibody responses.

Department

Description

Provenance

Citation

Published Version (Please cite this version)

10.1038/s41467-020-14670-w

Publication Info

Bradley, Todd, Masayuki Kuraoka, Chen-Hao Yeh, Ming Tian, Huan Chen, Derek W Cain, Xuejun Chen, Cheng Cheng, et al. (2020). Immune checkpoint modulation enhances HIV-1 antibody induction. Nature communications, 11(1). p. 948. 10.1038/s41467-020-14670-w Retrieved from https://hdl.handle.net/10161/21940.

This is constructed from limited available data and may be imprecise. To cite this article, please review & use the official citation provided by the journal.

Scholars@Duke

Yeh

Chen-Hao Yeh

Assistant Professor in Medicine

Dr. Yeh completed his undergraduate and Master of Science degree at the National Taiwan University in Taipei. He then pursued his Ph.D. at the University of Tokyo in Japan. He moved to Durham in 2015 for postdoctoral training in Dr. Garnett Kelsoe’s laboratory at the Duke Department of Immunology.

Dr. Yeh holds a broad academic background in biochemistry and immunology, with specific training and expertise in lymphocyte development and differentiation. His research has focused on: 1) germinal center (GC) B cell selection, differentiation and antibody affinity maturation and 2) T follicular helper (Tfh) cell differentiation and TCR repertoire analysis. 

Over the years, Dr. Yeh has demonstrated that B-cell selection based on surface pMHCII density is stringent in the establishment of GCs, but relatively relaxed during GC responses; this observation has led to fundamental revisions in the standard models for affinity-driven selection. With multiple genetic models to identify GC-resident Tfh cells in the mouse, Dr. Yeh also showed that the standard phenotypic definition of “GCTfh” included a majority of T cells that do not enter GCs. The more abundant Tfh-like cells have distinct developmental requirements, TCR repertoires and transcriptomic profiles compared to the rarer GC-resident Tfh cells, implying distinct physiologies and function. In addition, Dr. Yeh has categorized the phenotype of memory and GC B cell populations in Rhesus macaque (RM) as a step forward in understanding humoral responses in RMs and to enable isolation of live GC B cells for in vitro culture.

Cain

Derek Wilson Cain

Associate Professor in Medicine

My research focuses on the interactions of T cells and B cells during infection or following vaccination. I am particularly interested in the inter- and intracellular events that take place within germinal centers, the anatomic site of antibody evolution during an immune response.


Wiehe

Kevin J Wiehe

Associate Professor in Medicine

Dr. Kevin Wiehe is the director of research, director of computational biology and co-director of the Quantitative Research Division at the Duke Human Vaccine Institute (DHVI). He has over 20 years of experience in the field of computational biology and has expertise in computational structural biology, computational genomics, and computational immunology.

For the past decade, he has applied his unique background to developing computational approaches for studying the B cell response in both the infection and vaccination settings. He has utilized his expertise in computational structural biology to structurally model and characterize HIV and influenza antibody recognition. Dr. Wiehe has utilized his expertise in computational genomics and computational immunology to develop software to analyze large scale next generation sequencing data of antibody repertoires as well as develop computational programs for estimating antibody mutation probabilities. Dr. Wiehe has shown that low probability antibody mutations can act as rate-limiting steps in the development of broadly neutralizing antibodies in HIV.

Through his PhD, postdoc work, and now his roles at DHVI, Dr. Wiehe always approaches the analysis and the scientific discovery process from a structural biology perspective. Supporting the Duke Center for HIV Structural Biology (DCHSB), Dr. Wiehe will conduct antibody sequence analysis for antibodies used in computational and molecular modeling analyses conducted.

Saunders

Kevin O'Neil Saunders

Norman L. Letvin M. D. Distinguished Professor in Surgery and the Duke Human Vaccine Institute

Kevin O. Saunders, PhD, graduated from Davidson College in 2005 with a Bachelor of Science in biology. At Davidson College, he trained in the laboratory of Karen Hales, PhD, identifying the genetic basis of infertility. Saunders completed his doctoral research on CD8+ T cell immunity against HIV-1 infection with Georgia Tomaras, PhD, at Duke University in 2010. He subsequently trained as a postdoctoral fellow in the laboratories of Drs. Gary Nabel and John Mascola at the National Institutes of Health (NIH) National Institute of Allergy and Infectious Diseases (NIAID) Vaccine Research Center.

In 2014, Saunders joined the faculty at the Duke Human Vaccine Institute as a medical instructor. In this role, he analyzed antibody responses in vaccinated macaques, which led to the identification of glycan-dependent HIV antibodies induced by vaccination. Dr. Saunders was appointed as a non-tenure track assistant professor of surgery and the director of the laboratory of protein expression in the Duke Human Vaccine Institute in 2015. He successfully transitioned to a tenure-track appointment in 2018 and was later promoted to the rank of associate professor in surgery in 2020. In 2022, Saunders became an associate professor with tenure. He rose to the rank of professor with tenure in 2024 and was subsequently awarded the Norman L. Letvin, MD Professor in Immunology and Infectious Diseases Research in Surgery and the Duke Human Vaccine Institute distinguished professorship. Saunders previously served as DHVI's associate director of research, director or research, and currently serves as the associate director for DHVI. Additionally, Saunders serves as the faculty chairperson for DHVI's Diversity, Equity, and Inclusion Committee.

Saunders has given invited lectures at international conferences such as HIVR4P and the Keystone Symposia for HIV Vaccines. He has authored book chapters and numerous journal articles and holds patents on vaccine design concepts and antiviral antibodies. As a faculty member at Duke, Saunders has received the Duke Human Vaccine Institute Outstanding Leadership Award and the Norman Letvin Center For HIV/AIDS Vaccine Immunology and Immunogen Discovery Outstanding Investigator Award, Ruth and A. Morris Williams Faculty Research Prize, and the Duke Medical Alumni Emerging Leader Award. His current research interests include vaccine and antibody development to combat HIV-1, coronavirus, and other emerging viral infections.

About the Saunders Laboratory
The Saunders laboratory aims to understand the immunology of broadly protective antibodies and the molecular biology of their interaction with viral glycoprotein. The laboratory utilizes single B cell PCR, bulk B cell sequencing, and antigen-specific next-generation sequencing to probe the antibody repertoire during natural infection and after vaccination. The lab's overall goal is to develop protective antibody-based vaccines; therefore, the laboratory is divided into two sections–Immunoprofiling and Vaccine/Therapeutics design. They employ a reverse vaccinology approach to vaccine design where they study broadly protective antibodies in order to design vaccines that elicit such antibodies. To elicit broadly protective antibody responses, the Saunders laboratory utilizes epitope-focused nanoparticle vaccines. While eliciting broad protection is their overall goal, they are also interested in the immunologic mechanisms that make the vaccines successful.

Anti-glycan HIV-1 antibody biology. Their research premise is that vaccine-elicited antibodies will broadly neutralize HIV-1 if they can bind directly to the host glycans on Env. However, Env glycans are poorly immunogenic and require specific targeting by a vaccine immunogen to elicit an antibody response. Using this technique they identified two monoclonal antibodies from HIV Env vaccinated macaques called DH501 and DH502 that bind directly to mannose glycans and to HIV-1 envelope (Env). They have characterized these antibodies using glycan immunoassays, antibody engineering, and x-ray crystallography to define the mechanisms of Env-glycan interaction by these antibodies. Glycan-reactive HIV antibodies have mostly been found in the repertoire as IgG2 and IgM isotypes—similar to known natural glycan antibodies. Therefore they are examining whether vaccines mobilize antibodies from the natural glycan pool that affinity mature to interact with HIV-1 envelope. During this work, they discovered that Man9GlcNAc2 is the glycan preferred by early precursors in broadly neutralizing antibody lineages. They translated this finding into a vaccine design strategy that they have termed “glycan learning.” This approach modifies the number of glycans and type of glycosylation of HIV-1 Env immunogens to be optimal for engagement of the precursor antibody. The Env glycosylation sites and glycan type are then modified on subsequent Env immunogens to select antibodies that are maturing towards a broadly neutralizing phenotype. They have developed cell culture procedures and purification strategies combined with mass spectrometry analyses to create Env immunogens with specific glycosylation profiles. While the overall goal is to elicit protective neutralizing antibodies in vivo, they use these Env antigens in vitro to investigate the biology of B cell receptor engagement. 

HIV-1 Sequential vaccine design. The discovery of lineages of broadly neutralizing antibodies in HIV-infected individuals has provided templates for vaccine design. Utilizing viral sequences from individuals that make broadly neutralizing antibodies, we further engineer the viral protein to preferentially bind the desired type of antibody. The Saunders lab partners heavily with structural biologists and bioinformaticians to design optimized vaccine immunogens for in vitro and preclinical testing. They are investigating the hypothesis that broadly neutralizing antibodies can be engaged with envelope immunogens specifically designed to target them, and that engineered envelopes can select for the broadly neutralizing antibody precursors to develop into a broadly neutralizing antibody. They examine antibody responses in vaccinated humanized mice and monkeys to discern if the vaccine elicits antibodies that are similar to the known human broadly neutralizing antibody targets. Vaccines that are effective in animal models are translated for manufacturing and evaluation in Phase I clinical trials.

Pancoronavirus vaccine development. During the COVID-19 pandemic, the Saunders lab and DHVI as a whole worked to isolate broadly neutralizing antibodies against SARS-CoV-2 and related viruses. These antibodies then served as a template for the development of receptor binding domain nanoparticle vaccines we call RBD-scNP. These vaccines protected monkeys and mice from SARS-CoV-2 and animal coronaviruses. This vaccine has been translated to GMP manufacturing and will be examined in a Phase I clinical trial. The lab continues to apply similar approaches against other targets on coronaviruses to ultimately generate protective immunity against most coronaviruses. The lab explores different delivery methods including slow-release technology and nucleoside-modified mRNA delivery.

Taken together, our research program is an interdisciplinary approach to understanding the molecular biology underlying antibody recognition of viral glycoproteins in order to produce protective vaccines.


Unless otherwise indicated, scholarly articles published by Duke faculty members are made available here with a CC-BY-NC (Creative Commons Attribution Non-Commercial) license, as enabled by the Duke Open Access Policy. If you wish to use the materials in ways not already permitted under CC-BY-NC, please consult the copyright owner. Other materials are made available here through the author’s grant of a non-exclusive license to make their work openly accessible.