Efficient ex vivo expansion of conserved element vaccine-specific CD8+ T-cells from SHIV-infected, ART-suppressed nonhuman primates

HIV-specific T cells are necessary for control of HIV-1 replication but are largely insufficient for viral clearance. This is due in part to these cells' recognition of immunodominant but variable regions of the virus, which facilitates viral escape mutations that do not incur viral fitness cos...

Full description

Saved in:
Bibliographic Details
Published inFrontiers in immunology Vol. 14; p. 1188018
Main Authors Dross, Sandra, Venkataraman, Rasika, Patel, Shabnum, Huang, Meei-Li, Bollard, Catherine M, Rosati, Margherita, Pavlakis, George N, Felber, Barbara K, Bar, Katharine J, Shaw, George M, Jerome, Keith R, Mullins, James I, Kiem, Hans-Peter, Fuller, Deborah Heydenburg, Peterson, Christopher W
Format Journal Article
LanguageEnglish
Published Switzerland Frontiers Media S.A 03.05.2023
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:HIV-specific T cells are necessary for control of HIV-1 replication but are largely insufficient for viral clearance. This is due in part to these cells' recognition of immunodominant but variable regions of the virus, which facilitates viral escape mutations that do not incur viral fitness costs. HIV-specific T cells targeting conserved viral elements are associated with viral control but are relatively infrequent in people living with HIV (PLWH). The goal of this study was to increase the number of these cells an cell manufacturing approach derived from our clinically-validated HIV-specific expanded T-cell (HXTC) process. Using a nonhuman primate (NHP) model of HIV infection, we sought to determine i) the feasibility of manufacturing -expanded virus-specific T cells targeting viral conserved elements (CE, CE-XTCs), ii) the safety of these products, and iii) the impact of simian/human immunodeficiency virus (SHIV) challenge on their expansion, activity, and function. NHP CE-XTCs expanded up to 10-fold following co-culture with the combination of primary dendritic cells (DCs), PHA blasts pulsed with CE peptides, irradiated GM-K562 feeder cells, and autologous T cells from CE-vaccinated NHP. The resulting CE-XTC products contained high frequencies of CE-specific, polyfunctional T cells. However, consistent with prior studies with human HXTC and these cells' predominant CD8 effector phenotype, we did not observe significant differences in CE-XTC persistence or SHIV acquisition in two CE-XTC-infused NHP compared to two control NHP. These data support the safety and feasibility of our approach and underscore the need for continued development of CE-XTC and similar cell-based strategies to redirect and increase the potency of cellular virus-specific adaptive immune responses.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
Reviewed by: Joseph Mudd, Tulane University, United States; Mauricio A. Martins, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, United States; Ben Burwitz, Oregon Health and Science University, United States; Matthew S. Parsons, Armed Forces Research Institute of Medical Sciences, Thailand
Edited by: Ann Chahroudi, Emory University, United States
ISSN:1664-3224
1664-3224
DOI:10.3389/fimmu.2023.1188018